1
|
Liu H, Mei M, Lin S, Luo J, Huang S, Zhou J. Wuling San regulates AVPR2-cAMP-PKA-CREB pathway to delay cellular senescence and ameliorate acute kidney injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119679. [PMID: 40216046 DOI: 10.1016/j.jep.2025.119679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cellular senescence in renal resident cells plays a pivotal role in the progression of acute kidney injury (AKI), necessitating the expansion of effective drug targets. Traditional Chinese medicine (TCM) formulations, characterized by their multi-target effects, offer a promising perspective for advancing research on AKI. Wuling San (WLS), a well-established compound used in treating urological disorders, has yet to elucidate its potential pharmacological targets and mechanisms in ameliorating AKI and delaying cellular senescence. AIM OF THE STUDY This study sought to elucidate the mechanisms by which WLS modulates the AVPR2-cAMP-PKA-CREB pathway to mitigate cellular senescence and promote recovery from AKI. METHODS We first prepared WLS-containing serum and performed RT-qPCR experiments to screen for GPCRs that were differentially expressed in response to WLS. Next, we established an in vitro AKI mouse model to assess the renal protective effects of the WLS by measuring renal function, renal pathology, and oxidative stress levels. After this, we performed RNA sequencing (RNA-Seq) profiling to identify differentially expressed genes (DEGs) affected by WLS treatment. We also conducted Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses to identify potential signaling pathways involved. We then utilized the Gene Expression Omnibus (GEO) data to screen for cellular senescence related differentially expressed genes (CSRDEGs) in AKI patients and performed enrichment analysis, as well as a joint analysis of specific genes in relation to the RNA-Seq profiling results. We also examined how WLS affects the expression of proteins linked to cellular senescence in the AKI mouse model by targeting the AVPR2-cAMP-PKA-CREB pathway. RESULTS WLS markedly enhanced the expression of Arginine Vasopressin Receptor 2 (AVPR2) and ameliorated renal function indicators, as well as pathological changes and oxidative stress levels in the mouse model of AKI. RNA-Seq profiling revealed significant enrichment of the cAMP signaling pathway following WLS intervention. Bioinformatics analysis indicated that genes associated with cellular senescence in AKI patients were notably enriched in the p53 signaling pathway. Data mining from the GEO database, in conjunction with RNA-Seq profiling, demonstrated a substantial reduction in key genes after WLS treatment. Additionally, WLS elevated both the expression and phosphorylation of pivotal proteins within the AVPR2-cAMP-PKA-CREB pathway, while concurrently decreasing proteins associated with cellular senescence. CONCLUSION The results demonstrated that WLS significantly elevated the expression of AVPR2, which may underlie its nephroprotective effects and facilitate the mitigation of AKI by modulating the AVPR2-cAMP-PKA-CREB pathway, ultimately contributing to a delay in cellular senescence.
Collapse
Affiliation(s)
- Hong Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Manxue Mei
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Shuyin Lin
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Jiahui Luo
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Sirong Huang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
2
|
Gu X, Zhang Z, Zhao L, Lu L, Lu X, Li Y, Gu T, Huang X, Huang G, Liang Y, Meng H, Li B, Zhang X, Zhang J, Wang X, Du Y. Exposure to polyethylene terephthalate micro(nano)plastics exacerbates inflammation and fibrosis after myocardial infarction by reprogramming the gut and lung microbiota and metabolome. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137410. [PMID: 39919626 DOI: 10.1016/j.jhazmat.2025.137410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/06/2025] [Accepted: 01/25/2025] [Indexed: 02/09/2025]
Abstract
Micro(nano)plastics (MNPs), a ubiquitous environmental pollutant, have received increasing attention for their impacts on human health. We conducted an in-depth study on the role of polyethylene terephthalate (PET) MNPs in myocardial infarction (MI). Blood from the coronary circulation of MI patients was collected to detect microplastics (MPs). Peripheral monocytes (PBMCs) and AC16 cells were used to assess inflammation, cell proliferation and apoptosis after PET nanoplastics (NPs) stimulation. The mouse MI model was established after PET NPs respiratory or oral exposure. The results showed that various types of MPs, including high levels of PET MPs, were detected in the coronary circulation. PET NPs promoted inflammatory factors secretion by PBMCs, inhibited AC16 cell proliferation and promoted hypoxia-induced AC16 cell apoptosis. PET NPs exacerbated post-MI inflammation and fibrosis through activating the NLRP3 inflammasome pathway. Through macrogenetic sequencing and metabolomics analyses, we observed that PET NPs reprogrammed the intestinal and lung microbiota and metabolome in MI mice, leading to chronic inflammation. In conclusion, PET MPs were widely present in the coronary circulation of MI patients. PET MNPs can activate the NLRP3 inflammasome pathway to exacerbate post-MI ventricular remodelling, which may be related to the reprogramming of the gut and lung microbiota and metabolome.
Collapse
Affiliation(s)
- Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Zhixuan Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Lin Zhao
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Lijie Lu
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China
| | - Xin Lu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210004, China
| | - Yafei Li
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China
| | - Tianya Gu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xin Huang
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Guangyi Huang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Yan Liang
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Haoyu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Baihong Li
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Xiaodong Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Jun Zhang
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China.
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China.
| | - Yingqiang Du
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China.
| |
Collapse
|
3
|
Wang J, Hu S, Xu Y, Wang T. Omega-6 polyunsaturated fatty acids and their metabolites: a potential targeted therapy for pulmonary hypertension. Respir Res 2025; 26:102. [PMID: 40089708 PMCID: PMC11909876 DOI: 10.1186/s12931-025-03172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Pulmonary hypertension (PH) is a progressive and life-threatening cardiopulmonary disease that is not uncommon. The modulation of the pulmonary artery (PA) involves various fatty acids, including omega-6 polyunsaturated fatty acids (ω-6 PUFAs) and ω-6 PUFAs-derived oxylipins. These lipid mediators are produced through cyclooxygenase (COX), lipoxygenase (LOX), cytochrome P450 (CYP450), and non-enzymatic pathways. They play a crucial role in the occurrence and development of PH by regulating the function and phenotype of pulmonary artery endothelial cells (PAECs), pulmonary artery smooth muscle cells (PASMCs), pulmonary fibroblasts, alveolar macrophages, and inflammatory cells. The alterations in ω-6 PUFAs and oxylipins are pivotal in causing vasoconstriction, pulmonary remodeling, and ultimately leading to right heart failure in PH. Despite the limited understanding of the PH pathophysiology, there is potential for novel interventions through dietary and pharmacological approaches targeting ω-6 PUFAs and oxylipins. The aim of this review is to summarize the significant advances in clinical and basic research on omega-6 PUFAs and oxylipins in pulmonary vascular disease, particularly PH, and to propose a potential targeted therapeutic modality against omega-6 PUFAs.
Collapse
Affiliation(s)
- Jiayao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shunlian Hu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
4
|
Sparks BB, Ford H, Michelotti TC, Strieder-Barboza C. Adipose tissue oxylipin profile changes with subclinical ketosis and depot in postpartum dairy cows. J Dairy Sci 2025; 108:781-791. [PMID: 39343228 DOI: 10.3168/jds.2024-25178] [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: 05/16/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
Lipolysis of adipose tissue is a natural occurrence during the periparturient period in dairy cows. However, when lipolysis rates exceed the capacity of other tissues to used nonesterified fatty acids, it may lead to the development of ketosis and other diseases. Additionally, PUFA can become oxidized into oxylipins, which modulate inflammation and metabolism. The objective of this work was to identify depot-specific differences on adipose tissue oxylipin profile in dairy cows with and without subclinical ketosis and assess the effects of oxylipins on adipocyte function in vitro. Subcutaneous adipose tissue from the flank (SAT) and visceral adipose tissue from the omentum (VAT) were collected through laparotomy from multiparous dairy cows (5-14 DIM) and grouped according to blood BHB into nonketotic (NK; n = 5; BHB ≤ 0.8 mmol/L) and subclinical ketotic (SCK; n = 5; BHB 1.4 and ≤ 2.6 mmol/L). A targeted lipidome capable of detecting a 154 oxylipins was performed in paired SAT and VAT samples from all animals. Data were analyzed using the PROC GLIMMIX procedure in SAS (v9.4, SAS Institute Inc., Cary, NC) for the effect of depot (SAT, VAT), ketosis status (SCK, NK), and their interaction (depot × ketosis status) on oxylipin abundance. The oxylipins thromboxane-B2 (TXB2), prostaglandin-A2 (PGA2), and 5-hydroxeicostretanoic acid (5-HETE) were selected from lipidomic data based on effects of ketosis status and depot-specificity to further investigate their effects on SAT and VAT adipocyte function. Lipidomic data revealed 50 oxylipins across both adipose tissue depots. SCK was associated with a decreased abundance of TXB2 and tended to associate with an increase in PGA2 and prostaglandin-E1 (PGE1). Additionally, PGE1, 15-keto-prostaglandin-E2, 13,14-dihydro-15-keto-prostaglandin-E2, 5-HETE, and 15-HETE were increased in SAT. Although VAT had a greater abundance of 9,10-dihydroxy-12Z-octadecenoic acid, 12,13-dihydroxy-9Z-octadecenoic acid, 9-oxo-10E,12Z,15Z-octadecatrienoic acid, and 13S-hydroxy-9Z,11E,15Z-octadecatrienoic acid (13[s]-HOTrE). In vitro, an average (AVG) dose of 5-HETE on VAT cells tended to increase proliferation at d 7 compared with the control, HGH dose of TXB2 tended to decrease lipid accumulation in SAT compared with control, and AVG dose of PGA2 on VAT cells tended to lower ROS compared with the control. Overall, postpartum dairy cows have depot-specific adipose tissue lipidomic profiles which are associated with changes in ketosis status.
Collapse
Affiliation(s)
- Bridger B Sparks
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409
| | - Hunter Ford
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409
| | - Tainara C Michelotti
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409
| | - Clarissa Strieder-Barboza
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409; School of Veterinary Medicine, Texas Tech University, Amarillo, TX 79106.
| |
Collapse
|
5
|
Müller H, Hahn J, Gierke A, Stark R, Brunner C, Hoffmann TK, Greve J, Wittekindt O, Lochbaum R. Establishment of the deuterium oxide dilution method as a new possibility for determining the transendothelial water permeability. Pflugers Arch 2024; 476:993-1005. [PMID: 38438679 PMCID: PMC11139723 DOI: 10.1007/s00424-024-02934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/01/2024] [Accepted: 02/25/2024] [Indexed: 03/06/2024]
Abstract
Increase in transendothelial water permeability is an essential etiological factor in a variety of diseases like edema and shock. Despite the high clinical relevance, there has been no precise method to detect transendothelial water flow until now. The deuterium oxide (D2O) dilution method, already established for measuring transepithelial water transport, was used to precisely determine the transendothelial water permeability. It detected appropriate transendothelial water flow induced by different hydrostatic forces. This was shown in four different endothelial cell types. The general experimental setup was verified by gravimetry and absorbance spectroscopy. Determination of transendothelial electrical resistance (TEER) and immunocytochemical staining for proteins of the cell-cell contacts were performed to ensure that no damage to the endothelium occurred because of the measurements. Furthermore, endothelial barrier function was modulated. Measurement of transendothelial water flux was verified by measuring the TEER, the apparent permeability coefficient and the electrical capacity. The barrier-promoting substances cyclic adenosine monophosphate and iloprost reduced TEER and electrical capacity and increased permeability. This was accompanied by a reduced transendothelial water flux. In contrast, the barrier-damaging substances thrombin, histamine and bradykinin reduced TEER and electrical capacity, but increased permeability. Here, an increased water flow was shown. This newly established in vitro method for direct measurement of transendothelial water permeability was verified as a highly precise technique in various assays. The use of patient-specific endothelial cells enables individualized precision medicine in the context of basic edema research, for example regarding the development of barrier-protective pharmaceuticals.
Collapse
Affiliation(s)
- Hannes Müller
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - Janina Hahn
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - Angelina Gierke
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - Robert Stark
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - Thomas K Hoffmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - Jens Greve
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - Oliver Wittekindt
- Department of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Robin Lochbaum
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany.
| |
Collapse
|
6
|
Sun W, Wu W, Fang X, Ge X, Zhang Y, Han J, Guo X, Zhou L, Yang H. Disruption of pulmonary microvascular endothelial barrier by dysregulated claudin-8 and claudin-4: uncovered mechanisms in porcine reproductive and respiratory syndrome virus infection. Cell Mol Life Sci 2024; 81:240. [PMID: 38806818 PMCID: PMC11133251 DOI: 10.1007/s00018-024-05282-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
The pulmonary endothelium is a dynamic and metabolically active monolayer of endothelial cells. Dysfunction of the pulmonary endothelial barrier plays a crucial role in the acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), frequently observed in the context of viral pneumonia. Dysregulation of tight junction proteins can lead to the disruption of the endothelial barrier and subsequent leakage. Here, the highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) served as an ideal model for studying ALI and ARDS. The alveolar lavage fluid of pigs infected with HP-PRRSV, and the supernatant of HP-PRRSV infected pulmonary alveolar macrophages were respectively collected to treat the pulmonary microvascular endothelial cells (PMVECs) in Transwell culture system to explore the mechanism of pulmonary microvascular endothelial barrier leakage caused by viral infection. Cytokine screening, addition and blocking experiments revealed that proinflammatory cytokines IL-1β and TNF-α, secreted by HP-PRRSV-infected macrophages, disrupt the pulmonary microvascular endothelial barrier by downregulating claudin-8 and upregulating claudin-4 synergistically. Additionally, three transcription factors interleukin enhancer binding factor 2 (ILF2), general transcription factor III C subunit 2 (GTF3C2), and thyroid hormone receptor-associated protein 3 (THRAP3), were identified to accumulate in the nucleus of PMVECs, regulating the transcription of claudin-8 and claudin-4. Meanwhile, the upregulation of ssc-miR-185 was found to suppress claudin-8 expression via post-transcriptional inhibition. This study not only reveals the molecular mechanisms by which HP-PRRSV infection causes endothelial barrier leakage in acute lung injury, but also provides novel insights into the function and regulation of tight junctions in vascular homeostasis.
Collapse
Affiliation(s)
- Weifeng Sun
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- China Institute of Veterinary Drug Control, Beijing, 100081, People's Republic of China
| | - Weixin Wu
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Xinyu Fang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Xinna Ge
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Yongning Zhang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Jun Han
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Xin Guo
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Lei Zhou
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
| | - Hanchun Yang
- National Key Laboratory of Veterinary Public Health Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
- Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, People's Republic of China.
| |
Collapse
|
7
|
Shen X, Yang Z, Wang Q, Chen X, Zhu Q, Liu Z, Patel N, Liu X, Mo X. Lactobacillus plantarum L168 improves hyperoxia-induced pulmonary inflammation and hypoalveolarization in a rat model of bronchopulmonary dysplasia. NPJ Biofilms Microbiomes 2024; 10:32. [PMID: 38553470 PMCID: PMC10980738 DOI: 10.1038/s41522-024-00504-w] [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: 02/28/2023] [Accepted: 03/20/2024] [Indexed: 04/02/2024] Open
Abstract
Alteration of gut microbiota can affect chronic lung diseases, such as asthma and chronic obstructive pulmonary disease, through abnormal immune and inflammatory responses. Previous studies have shown a feasible connection between gut microbiota and bronchopulmonary dysplasia (BPD) in preterm infants. However, whether BPD can be ameliorated by restoring the gut microbiota remains unclear. In preterm infants with BPD, we found variance in the diversity and structure of gut microbiota. Similarly, BPD rats showed gut dysbiosis, characterized by a deficiency of Lactobacillus, which was abundant in normal rats. We therefore explored the effect and potential mechanism of action of a probiotic strain, Lactobacillus plantarum L168, in improving BPD. The BPD rats were treated with L. plantarum L168 by gavage for 2 weeks, and the effect was evaluated by lung histopathology, lung function, and serum inflammatory markers. Subsequently, we observed reduced lung injury and improved lung development in BPD rats exposed to L. plantarum L168. Further evaluation revealed that L. plantarum L168 improved intestinal permeability in BPD rats. Serum metabolomics showed altered inflammation-associated metabolites following L. plantarum L168 intervention, notably a marked increase in anti-inflammatory metabolites. In agreement with the metabolites analysis, RNA-seq analysis of the intestine and lung showed that inflammation and immune-related genes were down-regulated. Based on the information from RNA-seq, we validated that L. plantarum L168 might improve BPD relating to down-regulation of TLR4 /NF-κB /CCL4 pathway. Together, our findings suggest the potential of L. plantarum L168 to provide probiotic-based therapeutic strategies for BPD.
Collapse
Affiliation(s)
- Xian Shen
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaocong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Department of Neonatology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qihui Zhu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Zhi Liu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China
| | - Nishant Patel
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Pathogen of Jiangsu Province, Key Laboratory of Human Functional Genomics of Jiangsu Province Center of Global Health, Nanjing Medical University, Nanjing, China.
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Jia X, Huang J, Wu B, Yang M, Xu W. RNA-Seq profiling of circular RNAs in mice with lipopolysaccharide-induced acute lung injury. Genomics 2024; 116:110755. [PMID: 38061481 DOI: 10.1016/j.ygeno.2023.110755] [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: 07/13/2023] [Revised: 11/16/2023] [Accepted: 12/03/2023] [Indexed: 12/21/2023]
Abstract
Acute lung injury (ALI) is a serious illness that develops suddenly, progresses rapidly, has a poor treatment response and a high mortality rate. Studies have found that circular RNAs (circRNA) play a critical role in several diseases, but their role in ALI remains unclear. The aim of this study was to identify circRNAs that are associated with ALI and investigate their potential molecular mechanisms. A comparison of lung circRNA and microRNA expression profiles in mice with ALI and controls was performed by RNA-sequencing. A bioinformatic analysis was conducted to identify differentially expressed (DE) RNAs, to construct competitive endogenous RNA (ceRNA) networks, and to analyze their function and pathways. Then, a protein-protein interaction (PPI) network was generated by the Search Tool for the Retrieval of Interacting Genes database, and hub genes were identified using Cytoscape. Furthermore, a key ceRNA subnetwork was constructed based on these hub genes. Overall, we found 239 DE circRNAs and 42 DE microRNAs in ALI mice compared to controls. Additionally, the molecular mechanism of ALI was further understood by building ceRNA networks based on these DE genes. ALI-induced circRNAs are mostly function in the inflammatory response and metabolic processes. Moreover, DE circRNAs are primarily involved in the nuclear factor (NF)-kappa B and PI3K-Akt signaling pathways. Seven hub genes were derived from the PPI network of 191 genes, followed by the construction of circRNA-miRNA-hub gene subnetworks. In this study, circRNA profiles are remarkably changed in mice with LPS-triggered ALI, and their potential contribution to the disease is revealed.
Collapse
Affiliation(s)
- Xianxian Jia
- Department of Pediatrics, Shengjing Hospital of China Medical University, NO. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning Province, China
| | - Jinhui Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, NO. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning Province, China
| | - Bo Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, NO. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning Province, China
| | - Miao Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, NO. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning Province, China
| | - Wei Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, NO. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning Province, China.
| |
Collapse
|
9
|
Zhang W, Ding L, Zhang M, Zheng S, Ma R, Gong J, Mao H, Xu H, Xu P, Zhang Y. Dietary intake of α-ketoglutarate ameliorates α-synuclein pathology in mouse models of Parkinson's disease. Cell Mol Life Sci 2023; 80:155. [PMID: 37204481 PMCID: PMC11073026 DOI: 10.1007/s00018-023-04807-7] [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: 01/13/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
Parkinson's disease (PD) is a progressive movement disorder characterized by dopaminergic (DA) neuron degeneration and the existence of Lewy bodies formed by misfolded α-synuclein. Emerging evidence supports the benefits of dietary interventions in PD due to their safety and practicality. Previously, dietary intake of α-ketoglutarate (AKG) was proved to extend the lifespan of various species and protect mice from frailty. However, the mechanism of dietary AKG's effects in PD remains undetermined. In the present study, we report that an AKG-based diet significantly ameliorated α-synuclein pathology, and rescued DA neuron degeneration and impaired DA synapses in adeno-associated virus (AAV)-loaded human α-synuclein mice and transgenic A53T α-synuclein (A53T α-Syn) mice. Moreover, AKG diet increased nigral docosahexaenoic acid (DHA) levels and DHA supplementation reproduced the anti-α-synuclein effects in the PD mouse model. Our study reveals that AKG and DHA induced microglia to phagocytose and degrade α-synuclein via promoting C1q and suppressed pro-inflammatory reactions. Furthermore, results indicate that modulating gut polyunsaturated fatty acid metabolism and microbiota Lachnospiraceae_NK4A136_group in the gut-brain axis may underlie AKG's benefits in treating α-synucleinopathy in mice. Together, our findings propose that dietary intake of AKG is a feasible and promising therapeutic approach for PD.
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mengran Zhang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China
| | - Shaohui Zheng
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China
| | - Runfang Ma
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China
| | - Junwei Gong
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hengxu Mao
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Huaxi Xu
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Yunlong Zhang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, China.
| |
Collapse
|
10
|
Li L, Miao J, Shaheen N, Taleb SJ, Hu J, Ye Q, He J, Yan J, Mallampalli RK, Zhao J, Zhao Y. ISGylation of NF-κBp65 by SCF FBXL19 E3 Ligase Diminishes Endothelial Inflammation. Arterioscler Thromb Vasc Biol 2023; 43:674-683. [PMID: 36994728 PMCID: PMC10133096 DOI: 10.1161/atvbaha.122.318894] [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: 06/27/2022] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND NF-κB (nuclear factor kappa B) plays a pivotal role in endothelial cell (EC) inflammation. Protein ISGylation is regulated by E3 ISG15 (interferon-stimulated gene 15) ligases; however, ISGylation of NF-κBp65 and its role in EC functions have not been investigated. Here, we investigate whether p65 is ISGylated and the role of its ISGylation in endothelial functions. METHODS In vitro ISGylation assay and EC inflammation were performed. EC-specific transgenic mice were utilized in a murine model of acute lung injury. RESULTS We find that NF-κBp65 is ISGylated in resting ECs and that the posttranslational modification is reversible. TNFα (tumor necrosis factor alpha) and endotoxin stimulation of EC reduce p65 ISGylation, promoting its serine phosphorylation through reducing its association with a phosphatase WIP1 (wild-type p53-induced phosphatase 1). Mechanistically, an SCF (Skp1-Cul1-F-box) protein E3 ligase SCFFBXL19 is identified as a new ISG15 E3 ligase that targets and catalyzes ISGylation of p65. Depletion of FBXL19 (F-box and leucine-rich repeat protein 19) increases p65 phosphorylation and EC inflammation, suggesting a negative correlation between p65 ISGylation and phosphorylation. Moreover, EC-specific FBXL19 overexpressing humanized transgenic mice exhibit reduced lung inflammation and severity of experimental acute lung injury. CONCLUSIONS Together, our data reveal a new posttranslational modification of p65 catalyzed by a previously unrecognized role of SCFFBXL19 as an ISG15 E3 ligase that modulates EC inflammation.
Collapse
Affiliation(s)
- Lian Li
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
| | - Jiaxing Miao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
| | - Nargis Shaheen
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
| | - Sarah J. Taleb
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
| | - Jian Hu
- Department of Internal Medicine, the Ohio State University, Columbus, OH
| | - Qinmao Ye
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
| | - Jinshan He
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
| | - Jiasheng Yan
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
| | | | - Jing Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
- Department of Internal Medicine, the Ohio State University, Columbus, OH
| | - Yutong Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University, Columbus, OH
- Department of Internal Medicine, the Ohio State University, Columbus, OH
| |
Collapse
|
11
|
Sun Z, Chen X, Liu J, Du Y, Duan C, Xiao S, Zhou Y, Fang L. PRRSV-induced inflammation in pulmonary intravascular macrophages (PIMs) and pulmonary alveolar macrophages (PAMs) contributes to endothelial barrier function injury. Vet Microbiol 2023; 281:109730. [PMID: 37068404 DOI: 10.1016/j.vetmic.2023.109730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/28/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a severe infectious disease currently devasting the global pig industry. PRRS is characterized by intense inflammation and severe damage to the alveolar-capillary barrier. Therefore, it is crucial to uncover the underlying mechanism by which the PRRS virus (PRRSV) induces inflammatory responses and barrier function damage. In addition to porcine alveolar macrophages (PAMs), the primary target cells of PRRSV infection in vivo, pulmonary intravascular macrophages (PIMs) are also susceptible to PRRSV infection. However, the poor isolation efficiency limits the study of PRRSV infection in PIMs. In this study, we optimized the isolation method to obtain PIMs with higher purity and yield and demonstrated that PRRSV's infection kinetics in PIMs were similar to those in PAMs. Notably, PIMs exhibited a more acute inflammation process during PRRSV infection than PAMs, as evidenced by the earlier upregulation and higher levels of pro-inflammatory cytokines, including TNF-α and IL-1β. More acute endothelial barrier disfunction upon PRRSV infection was also observed in PIMs compared to in PAMs. Mechanistically, PRRSV-induced TNF-α and IL-1β could cause endothelial barrier disfunction by dysregulating tight junction proteins, including claudin 1 (CLDN1), claudin 8 (CLDN8) and occludin (OCLN). Our findings revealed the crucial and novel roles of PIMs in facilitating the progression of inflammatory responses and endothelial barrier injury and provided new insights into the mechanisms of PRRSV's induction of interstitial pneumonia.
Collapse
|
12
|
Significance of Pulmonary Endothelial Injury and the Role of Cyclooxygenase-2 and Prostanoid Signaling. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010117. [PMID: 36671689 PMCID: PMC9855370 DOI: 10.3390/bioengineering10010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The endothelium plays a key role in the dynamic balance of hemodynamic, humoral and inflammatory processes in the human body. Its central importance and the resulting therapeutic concepts are the subject of ongoing research efforts and form the basis for the treatment of numerous diseases. The pulmonary endothelium is an essential component for the gas exchange in humans. Pulmonary endothelial dysfunction has serious consequences for the oxygenation and the gas exchange in humans with the potential of consecutive multiple organ failure. Therefore, in this review, the dysfunction of the pulmonary endothel due to viral, bacterial, and fungal infections, ventilator-related injury, and aspiration is presented in a medical context. Selected aspects of the interaction of endothelial cells with primarily alveolar macrophages are reviewed in more detail. Elucidation of underlying causes and mechanisms of damage and repair may lead to new therapeutic approaches. Specific emphasis is placed on the processes leading to the induction of cyclooxygenase-2 and downstream prostanoid-based signaling pathways associated with this enzyme.
Collapse
|
13
|
Godbole NM, Chowdhury AA, Chataut N, Awasthi S. Tight Junctions, the Epithelial Barrier, and Toll-like Receptor-4 During Lung Injury. Inflammation 2022; 45:2142-2162. [PMID: 35779195 PMCID: PMC9649847 DOI: 10.1007/s10753-022-01708-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/31/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022]
Abstract
Lung epithelium is constantly exposed to the environment and is critically important for the orchestration of initial responses to infectious organisms, toxins, and allergic stimuli, and maintenance of normal gaseous exchange and pulmonary function. The integrity of lung epithelium, fluid balance, and transport of molecules is dictated by the tight junctions (TJs). The TJs are formed between adjacent cells. We have focused on the topic of the TJ structure and function in lung epithelial cells. This review includes a summary of the last twenty years of literature reports published on the disrupted TJs and epithelial barrier in various lung conditions and expression and regulation of specific TJ proteins against pathogenic stimuli. We discuss the molecular signaling and crosstalk among signaling pathways that control the TJ structure and function. The Toll-like receptor-4 (TLR4) recognizes the pathogen- and damage-associated molecular patterns released during lung injury and inflammation and coordinates cellular responses. The molecular aspects of TLR4 signaling in the context of TJs or the epithelial barrier are not fully known. We describe the current knowledge and possible networking of the TLR4-signaling with cellular and molecular mechanisms of TJs, lung epithelial barrier function, and resistance to treatment strategies.
Collapse
Affiliation(s)
- Nachiket M Godbole
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Asif Alam Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Neha Chataut
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Shanjana Awasthi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
14
|
Inhibition of Lipopolysaccharide-Induced Inflammatory Signaling by Soft Coral-Derived Prostaglandin A 2 in RAW264.7 Cells. Mar Drugs 2022; 20:md20050316. [PMID: 35621967 PMCID: PMC9143063 DOI: 10.3390/md20050316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
Lipopolysaccharide (LPS) is a component of the outer membrane of Gram-negative bacteria and causes inflammatory diseases. We searched MeOH extracts of collected marine organisms for inhibitors of LPS-induced nitric oxide (NO) production in RAW264.7 cells and identified prostaglandin A2 (PGA2) as an active compound from the MeOH extract of the soft coral Lobophytum sp. PGA2 inhibited the production of NO and reduced the expression of inducible NO synthase (iNOS) in LPS-stimulated RAW264.7 cells. Although short preincubation with PGA2 did not inhibit LPS-induced degradation and resynthesis of IκBα, the suppressive effect of PGA2 was observed only after a prolonged incubation period prior to LPS treatment. In addition, PGA2-inhibited NO production was negated by the addition of the EP4 antagonist L161982. Thus, PGA2 was identified as an inhibitor of LPS-induced inflammatory signaling in RAW264.7 cells.
Collapse
|
15
|
Bandela M, Belvitch P, Garcia JGN, Dudek SM. Cortactin in Lung Cell Function and Disease. Int J Mol Sci 2022; 23:4606. [PMID: 35562995 PMCID: PMC9101201 DOI: 10.3390/ijms23094606] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/30/2022] Open
Abstract
Cortactin (CTTN) is an actin-binding and cytoskeletal protein that is found in abundance in the cell cortex and other peripheral structures of most cell types. It was initially described as a target for Src-mediated phosphorylation at several tyrosine sites within CTTN, and post-translational modifications at these tyrosine sites are a primary regulator of its function. CTTN participates in multiple cellular functions that require cytoskeletal rearrangement, including lamellipodia formation, cell migration, invasion, and various other processes dependent upon the cell type involved. The role of CTTN in vascular endothelial cells is particularly important for promoting barrier integrity and inhibiting vascular permeability and tissue edema. To mediate its functional effects, CTTN undergoes multiple post-translational modifications and interacts with numerous other proteins to alter cytoskeletal structures and signaling mechanisms. In the present review, we briefly describe CTTN structure, post-translational modifications, and protein binding partners and then focus on its role in regulating cellular processes and well-established functional mechanisms, primarily in vascular endothelial cells and disease models. We then provide insights into how CTTN function affects the pathophysiology of multiple lung disorders, including acute lung injury syndromes, COPD, and asthma.
Collapse
Affiliation(s)
- Mounica Bandela
- Department of Biomedical Engineering, College of Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Patrick Belvitch
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Joe G. N. Garcia
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA;
| | - Steven M. Dudek
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
16
|
Highly Pathogenic PRRSV-Infected Alveolar Macrophages Impair the Function of Pulmonary Microvascular Endothelial Cells. Viruses 2022; 14:v14030452. [PMID: 35336858 PMCID: PMC8948932 DOI: 10.3390/v14030452] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/13/2023] Open
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV), especially the highly pathogenic strains, can cause serious acute lung injury (ALI), characterized by extensive hemorrhage, inflammatory cells and serous fluid infiltration in the lung vascular system. Meanwhile, the pulmonary microvascular endothelial cells (PMVECs) are essential for forming the air–blood barrier and keeping the water–salt balance to prevent leakage of circulating nutrients, solutes, and fluid into the underlying tissues. As well, they tightly regulate the influx of immune cells. To determine the possible relationship between the PMVECs’ function changes and lung vascular permeability during PRRSV infection, the PMVECs were co-cultured with HP-PRRSV-inoculated primary pulmonary alveolar macrophages (PAMs) in transwell model, and then the RNA sequencing (RNA-seq) and comprehensive bioinformatics analysis were carried out to characterize the dynamic transcriptome landscapes of PMVECs. In total, 16,489 annotated genes were identified, with 275 upregulated and 270 downregulated differentially expressed genes (DEGs) were characterized at both 18 and 24 h post PRRSV inoculation. The GO terms and KEGG pathways analysis indicated that the immune response, metabolic pathways, cell death, cytokine–cytokine receptor interaction, viral responses, and apoptotic process are significantly regulated upon co-culture with PRRSV-infected PAMs. Moreover, according to the TERR and dextran flux assay results, dysregulation of TJ proteins, including CLDN1, CLDN4, CLDN8, and OCLN, is further confirmed to correlate with the increased permeability of PMVECs. These transcriptome profiles and DEGs will provide valuable clues for further exploring the roles of PMVECs in PRRSV-induced ALI in the future.
Collapse
|
17
|
Yamamoto K, Takagi Y, Ando K, Fukuhara S. Rap1 Small GTPase Regulates Vascular Endothelial-Cadherin-Mediated Endothelial Cell-Cell Junctions and Vascular Permeability. Biol Pharm Bull 2021; 44:1371-1379. [PMID: 34602545 DOI: 10.1248/bpb.b21-00504] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The vascular permeability of the endothelium is finely controlled by vascular endothelial (VE)-cadherin-mediated endothelial cell-cell junctions. In the majority of normal adult tissues, endothelial cells in blood vessels maintain vascular permeability at a relatively low level, while in response to inflammation, they limit vascular barrier function to induce plasma leakage and extravasation of immune cells as a defense mechanism. Thus, the dynamic but also simultaneously tight regulation of vascular permeability by endothelial cells is responsible for maintaining homeostasis and, as such, impairments of its underlying mechanisms result in hyperpermeability, leading to the development and progression of various diseases including coronavirus disease 2019 (COVID-19), a newly emerging infectious disease. Recently, increasing numbers of studies have been unveiling the important role of Rap1, a small guanosine 5'-triphosphatase (GTPase) belonging to the Ras superfamily, in the regulation of vascular permeability. Rap1 enhances VE-cadherin-mediated endothelial cell-cell junctions to potentiate vascular barrier functions via dynamic reorganization of the actin cytoskeleton. Importantly, Rap1 signaling activation reportedly improves vascular barrier function in animal models of various diseases associated with vascular hyperpermeability, suggesting that Rap1 might be an ideal target for drugs intended to prevent vascular barrier dysfunction. Here, we describe recent progress in understanding the mechanisms by which Rap1 potentiates VE-cadherin-mediated endothelial cell-cell adhesions and vascular barrier function. We also discuss how alterations in Rap1 signaling are related to vascular barrier dysfunction in diseases such as acute pulmonary injury and malignancies. In addition, we examine the possibility of Rap1 signaling as a target of drugs for treating diseases associated with vascular hyperpermeability.
Collapse
Affiliation(s)
- Kiyotake Yamamoto
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| | - Yuki Takagi
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| | - Koji Ando
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| |
Collapse
|
18
|
Xu Q, Chen J, Zhu Y, Xia W, Liu Y, Xu J. JTE-013 Alleviates Inflammatory Injury and Endothelial Dysfunction Induced by Sepsis In Vivo and In Vitro. J Surg Res 2021; 265:323-332. [PMID: 33971464 DOI: 10.1016/j.jss.2021.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 02/09/2021] [Accepted: 03/03/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Nowadays, there is no approved targeted agent for lung injury induced by sepsis. S1PR2 is confirmed to be a promising diagnosis and treatment target. JTE-013 as S1PR2 antagonists may be an agent of great potential. In this research, we sought to determine the functional role of JTE-013 in lung injury induced by sepsis. MATERIALS AND METHODS Seventy-two rats were assigned into normal group, sepsis model group and JTE-013 group. The animal model of lung injury induced by sepsis was constructed by cecal ligation and puncture. The human pulmonary microvascular endothelial cells (HPMECs) were divided into control, LPS and LPS + JTE-013 group. HPMECs induced by LPS served as the cell model of lung injury induced by sepsis. HE staining assay was performed for assessment of the pathological condition and Evans blue was applied for assessment of pulmonary tissue permeability. Wet/dry ratio was measured as indicators of pulmonary edema degree and neutrophil count was measured as indicators of infection status. The levels of inflammatory factors were detected by corresponding kits, cell survival by CCK-8 assay and protein expression level by western blot. RESULTS S1PR2 was highly expressed in vivo model of lung injury induced by sepsis. It was observed that JTE-013 as antagonist of S1PR2 alleviated the lung tissue injury, endothelial dysfunction and pulmonary edema induced by sepsis. In addition, JTE-013 reduced neutrophil count and levels of inflammatory factors. Moreover, results confirmed that JTE-013 enhanced cell viability and mitigated inflammatory response in cell model of sepsis. CONCLUSIONS Overall, JTE-013 as an antagonist of S1PR2 could relieve inflammatory injury and endothelial dysfunction induced by sepsis in vivo and vitro, resulting in attenuation of lung injury. These findings elucidated that JTE-013 may be a promising targeted agent for lung injury induced by sepsis.
Collapse
Affiliation(s)
- Qiumin Xu
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Jiusheng Chen
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Yifan Zhu
- Convalescent Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Wenjuan Xia
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Yong Liu
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Jieying Xu
- Special Functions Section, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China.
| |
Collapse
|
19
|
Liu K, Mao W, Liu B, Li T, Wang X, Pei L, Cao J, Wang F. Prostaglandin E2 promotes Staphylococcus aureus infection via EP4 receptor in bovine endometrium. Microb Pathog 2021; 158:105019. [PMID: 34107344 DOI: 10.1016/j.micpath.2021.105019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 10/21/2022]
Abstract
Prostaglandin E2 (PGE2) enhances Staphylococcus aureus infection but its mechanism is not well understood. Here, we examined the effect of PGE2 on Staphylococcal Protein A (SPA) expression in bovine endometrium and determined the role of select PGE2 receptors (i.e., EP2 and EP4) in adhesion and internalization of S. aureus. S. aureus isolate SA113 was used for in vitro infection of bovine endometrial tissues and epithelial cells, with treatment conditions consisting of untreated control, SA113 treatment, SA113 + PGE2, SA113 + PGE2 + EP2 receptor antagonist (AH-6809), and SA113 + PGE2 + EP4 receptor antagonist (AH-23848). Immunofluorescence assay revealed that PGE2 could promote SPA expression in S. aureus-infected bovine endometrial tissues. PGE2 also enhanced the adhesion and internalization of S. aureus in bovine endometrial cells. The addition of EP4 antagonist, but not the EP2 antagonist, abrogated the ability of PGE2 to promote S. aureus SPA expression, adhesion, and internalization in endometrial cells. Our findings suggest that S. aureus infection in the endometrium is enhanced by PGE2 through the EP4 receptor. This result is essential for the development of new approach to treating S. aureus infection, such as the application of EP4 antagonist as an adjunct drug treatment.
Collapse
Affiliation(s)
- Kun Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China; Laboratory of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China.
| | - Wei Mao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Bo Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Tingting Li
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Xinfei Wang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Le Pei
- Veterinary Research Institute, Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot, China.
| | - Jinshan Cao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, 010018, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Anima Disease, Ministry of Agriculture, Hohhot, China.
| | - Fenglong Wang
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
20
|
Kim YM, Krantz S, Jambusaria A, Toth PT, Moon HG, Gunarathna I, Park GY, Rehman J. Mitofusin-2 stabilizes adherens junctions and suppresses endothelial inflammation via modulation of β-catenin signaling. Nat Commun 2021; 12:2736. [PMID: 33980844 PMCID: PMC8115264 DOI: 10.1038/s41467-021-23047-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/13/2021] [Indexed: 01/19/2023] Open
Abstract
Endothelial barrier integrity is ensured by the stability of the adherens junction (AJ) complexes comprised of vascular endothelial (VE)-cadherin as well as accessory proteins such as β-catenin and p120-catenin. Disruption of the endothelial barrier due to disassembly of AJs results in tissue edema and the influx of inflammatory cells. Using three-dimensional structured illumination microscopy, we observe that the mitochondrial protein Mitofusin-2 (Mfn2) co-localizes at the plasma membrane with VE-cadherin and β-catenin in endothelial cells during homeostasis. Upon inflammatory stimulation, Mfn2 is sulfenylated, the Mfn2/β-catenin complex disassociates from the AJs and Mfn2 accumulates in the nucleus where Mfn2 negatively regulates the transcriptional activity of β-catenin. Endothelial-specific deletion of Mfn2 results in inflammatory activation, indicating an anti-inflammatory role of Mfn2 in vivo. Our results suggest that Mfn2 acts in a non-canonical manner to suppress the inflammatory response by stabilizing cell-cell adherens junctions and by binding to the transcriptional activator β-catenin.
Collapse
Affiliation(s)
- Young-Mee Kim
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Sarah Krantz
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ankit Jambusaria
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Peter T Toth
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Research Resources Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Isuru Gunarathna
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
21
|
Vivarelli S, Falzone L, Torino F, Scandurra G, Russo G, Bordonaro R, Pappalardo F, Spandidos DA, Raciti G, Libra M. Immune-checkpoint inhibitors from cancer to COVID‑19: A promising avenue for the treatment of patients with COVID‑19 (Review). Int J Oncol 2021; 58:145-157. [PMID: 33491759 PMCID: PMC7864014 DOI: 10.3892/ijo.2020.5159] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
The severe acute respiratory syndrome associated coronavirus‑2 (SARS‑CoV‑2) poses a threat to human life worldwide. Since early March, 2020, coronavirus disease 2019 (COVID‑19), characterized by an acute and often severe form of pneumonia, has been declared a pandemic. This has led to a boom in biomedical research studies at all stages of the pipeline, from the in vitro to the clinical phase. In line with this global effort, known drugs, currently used for the treatment of other pathologies, including antivirals, immunomodulating compounds and antibodies, are currently used off‑label for the treatment of COVID‑19, in association with the supportive standard care. Yet, no effective treatments have been identified. A new hope stems from medical oncology and relies on the use of immune‑checkpoint inhibitors (ICIs). In particular, amongst the ICIs, antibodies able to block the programmed death‑1 (PD‑1)/PD ligand-1 (PD‑L1) pathway have revealed a hidden potential. In fact, patients with severe and critical COVID‑19, even prior to the appearance of acute respiratory distress syndrome, exhibit lymphocytopenia and suffer from T‑cell exhaustion, which may lead to viral sepsis and an increased mortality rate. It has been observed that cancer patients, who usually are immunocompromised, may restore their anti‑tumoral immune response when treated with ICIs. Moreover, viral-infected mice and humans, exhibit a T‑cell exhaustion, which is also observed following SARS‑CoV‑2 infection. Importantly, when treated with anti‑PD‑1 and anti‑PD‑L1 antibodies, they restore their T‑cell competence and efficiently counteract the viral infection. Based on these observations, four clinical trials are currently open, to examine the efficacy of anti‑PD‑1 antibody administration to both cancer and non‑cancer individuals affected by COVID‑19. The results may prove the hypothesis that restoring exhausted T‑cells may be a winning strategy to beat SARS‑CoV‑2 infection.
Collapse
Affiliation(s)
- Silvia Vivarelli
- Section of General Pathology, Clinics and Oncology, Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania
| | - Luca Falzone
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori 'Fondazione G. Pascale', I-80131 Naples
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, I-00133 Rome
| | | | - Giulia Russo
- Department of Drug Sciences, University of Catania, I-95123 Catania
| | | | - Francesco Pappalardo
- Department of Drug Sciences, University of Catania, I-95123 Catania
- Research Center for Prevention, Diagnosis and Treatment of Tumors, University of Catania, I-95123 Catania, Italy
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Massimo Libra
- Section of General Pathology, Clinics and Oncology, Department of Biomedical and Biotechnological Sciences, University of Catania, I-95123 Catania
- Research Center for Prevention, Diagnosis and Treatment of Tumors, University of Catania, I-95123 Catania, Italy
| |
Collapse
|
22
|
Karki P, Birukov KG. Oxidized Phospholipids in Control of Endothelial Barrier Function: Mechanisms and Implication in Lung Injury. Front Endocrinol (Lausanne) 2021; 12:794437. [PMID: 34887839 PMCID: PMC8649713 DOI: 10.3389/fendo.2021.794437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/05/2021] [Indexed: 01/25/2023] Open
Abstract
Earlier studies investigating the pathogenesis of chronic vascular inflammation associated with atherosclerosis described pro-inflammatory and vascular barrier disruptive effects of lipid oxidation products accumulated in the sites of vascular lesion and atherosclerotic plaque. However, accumulating evidence including studies from our group suggests potent barrier protective and anti-inflammatory properties of certain oxidized phospholipids (OxPLs) in the lung vascular endothelium. Among these OxPLs, oxidized 1-palmitoyl-2-arachdonyl-sn-glycero-3-phosphocholine (OxPAPC) causes sustained enhancement of lung endothelial cell (EC) basal barrier properties and protects against vascular permeability induced by a wide variety of agonists ranging from bacterial pathogens and their cell wall components, endotoxins, thrombin, mechanical insults, and inflammatory cytokines. On the other hand, truncated OxPLs cause acute endothelial barrier disruption and potentiate inflammation. It appears that multiple signaling mechanisms triggering cytoskeletal remodeling are involved in OxPLs-mediated regulation of EC barrier. The promising vascular barrier protective and anti-inflammatory properties exhibited by OxPAPC and its particular components that have been established in the cellular and animal models of sepsis and acute lung injury has prompted consideration of OxPAPC as a prototype therapeutic molecule. In this review, we will summarize signaling and cytoskeletal mechanisms involved in OxPLs-mediated damage, rescue, and restoration of endothelial barrier in various pathophysiological settings and discuss a future potential of OxPAPC in treating lung disorders associated with endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: Konstantin G. Birukov,
| |
Collapse
|
23
|
Maiuolo J, Mollace R, Gliozzi M, Musolino V, Carresi C, Paone S, Scicchitano M, Macrì R, Nucera S, Bosco F, Scarano F, Zito MC, Ruga S, Tavernese A, Mollace V. The Contribution of Endothelial Dysfunction in Systemic Injury Subsequent to SARS-Cov-2 Infection. Int J Mol Sci 2020; 21:E9309. [PMID: 33291346 PMCID: PMC7730352 DOI: 10.3390/ijms21239309] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) infection is associated, alongside with lung infection and respiratory disease, to cardiovascular dysfunction that occurs at any stage of the disease. This includes ischemic heart disease, arrhythmias, and cardiomyopathies. The common pathophysiological link between SARS-CoV-2 infection and the cardiovascular events is represented by coagulation abnormalities and disruption of factors released by endothelial cells, which contribute in maintaining the blood vessels into an anti-thrombotic state. Thus, early alteration of the functionality of endothelial cells, which may be found soon after SARS-CoV-2 infection, seems to represent the major target of a SARS CoV-2 disease state and accounts for the systemic vascular dysfunction that leads to a detrimental effect in terms of hospitalization and death accompanying the disease. In particular, the molecular interaction of SARS-CoV-2 with the ACE2 receptor located in the endothelial cell surface, either at the pulmonary and systemic level, leads to early impairment of endothelial function, which, in turn, is followed by vascular inflammation and thrombosis of peripheral blood vessels. This highlights systemic hypoxia and further aggravates the vicious circle that compromises the development of the disease, leading to irreversible tissue damage and death of people with SARS CoV-2 infection. The review aims to assess some recent advances to define the crucial role of endothelial dysfunction in the pathogenesis of vascular complications accompanying SARS-CoV-2 infection. In particular, the molecular mechanisms associated with the interaction of SARS CoV-2 with the ACE2 receptor located on the endothelial cells are highlighted to support its role in compromising endothelial cell functionality. Finally, the consequences of endothelial dysfunction in enhancing pro-inflammatory and pro-thrombotic effects of SARS-CoV-2 infection are assessed in order to identify early therapeutic interventions able to reduce the impact of the disease in high-risk patients.
Collapse
Affiliation(s)
- Jessica Maiuolo
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Rocco Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Department of Medicine, Chair of Cardiology, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Sara Paone
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
| | - Roberta Macrì
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
| | - Stefano Ruga
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
| | - Annamaria Tavernese
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Department of Medicine, Chair of Cardiology, University of Rome Tor Vergata, 00133 Roma, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (J.M.); (R.M.); (M.G.); (V.M.); (C.C.); (S.P.); (M.S.); (R.M.); (S.N.); (F.B.); (F.S.); (M.C.Z.); (S.R.); (A.T.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele Pisana, 00163 Roma, Italy
| |
Collapse
|
24
|
Du Y, Taylor CG, Aukema HM, Zahradka P. Role of oxylipins generated from dietary PUFAs in the modulation of endothelial cell function. Prostaglandins Leukot Essent Fatty Acids 2020; 160:102160. [PMID: 32717531 DOI: 10.1016/j.plefa.2020.102160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/17/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Abstract
Oxylipins, which are circulating bioactive lipids generated from polyunsaturated fatty acids (PUFAs) by cyclooxygenase, lipooxygenase and cytochrome P450 enzymes, have diverse effects on endothelial cells. Although studies of the effects of oxylipins on endothelial cell function are accumulating, a review that provides a comprehensive compilation of current knowledge and recent advances in the context of vascular homeostasis is lacking. This is the first compilation of the various in vitro, ex vivo and in vivo reports to examine the effects and potential mechanisms of action of oxylipins on endothelial cells. The aggregate data indicate docosahexaenoic acid-derived oxylipins consistently show beneficial effects related to key endothelial cell functions, whereas oxylipins derived from other PUFAs exhibit both positive and negative effects. Furthermore, information is lacking for certain oxylipin classes, such as those derived from α-linolenic acid, which suggests additional studies are required to achieve a full understanding of how oxylipins affect endothelial cells.
Collapse
Affiliation(s)
- Youjia Du
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada
| | - Carla G Taylor
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada
| | - Harold M Aukema
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada
| | - Peter Zahradka
- Canadian Centre for Agri-Food Research in Health and Medicine, St Boniface Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada; Department of Physiology and Pathophysiology, University of Manitoba, MB R3E 0J9, Canada; Department of Food and Human Nutritional Sciences, University of Manitoba, MB R3T 2N2, Canada.
| |
Collapse
|
25
|
Junaid A, Schoeman J, Yang W, Stam W, Mashaghi A, van Zonneveld AJ, Hankemeier T. Metabolic response of blood vessels to TNFα. eLife 2020; 9:54754. [PMID: 32749215 PMCID: PMC7476757 DOI: 10.7554/elife.54754] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 08/02/2020] [Indexed: 12/11/2022] Open
Abstract
TNFα signaling in the vascular endothelium elicits multiple inflammatory responses that drive vascular destabilization and leakage. Bioactive lipids are main drivers of these processes. In vitro mechanistic studies of bioactive lipids have been largely based on two-dimensional endothelial cell cultures that, due to lack of laminar flow and the growth of the cells on non-compliant stiff substrates, often display a pro-inflammatory phenotype. This complicates the assessment of inflammatory processes. Three-dimensional microvessels-on-a-chip models provide a unique opportunity to generate endothelial microvessels in a more physiological environment. Using an optimized targeted liquid chromatography–tandem mass spectrometry measurements of a panel of pro- and anti-inflammatory bioactive lipids, we measure the profile changes upon administration of TNFα. We demonstrate that bioactive lipid profiles can be readily detected from three-dimensional microvessels-on-a-chip and display a more dynamic, less inflammatory response to TNFα, that resembles more the human situation, compared to classical two-dimensional endothelial cell cultures. In a range of conditions called autoimmune diseases, the immune system attacks the body rather than foreign elements. This can cause inflammation that is harmful for many organs. In particular, immune cells can produce excessive amounts of a chemical messenger called tumor necrosis factor alpha (TNFα for short), which can lead to the release of fatty molecules that damage blood vessels. This process is normally studied in blood vessels cells that are grown on a dish, without any blood movement. However, in this rigid 2D environment, the cells become ‘stressed’ and show higher levels of inflammation than in the body. This makes it difficult to assess the exact role that TNFα plays in disease. A new technology is addressing this issue by enabling scientist to culture blood vessels cells in dishes coated with gelatin. This allows the cells to organize themselves in 3D, creating tiny blood vessels in which fluids can flow. However, it was unclear whether these ‘microvessels-on-a-chip’ were better models to study the role of TNFα compared to cells grown on a plate. Here, Junaid et al. compared the levels of inflammation in blood vessels cells grown in the two environments, showing that cells are less inflamed when they are cultured in 3D. In addition, when the artificial 3D-blood vessels were exposed to TNFα, they responded more like real blood vessels than the 2D models. Finally, experiments showed that it was possible to monitor the release of fatty molecules in this environment. Together, this work suggests that microvessels-on-a-chip are better models to study how TNFα harms blood vessels. Next, systems and protocols could be develop to allow automated mass drug testing in microvessels-on-a-chip. This would help scientists to quickly screen thousands of drugs and find candidates that can protect blood vessels from TNFα.
Collapse
Affiliation(s)
- Abidemi Junaid
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands.,Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Johannes Schoeman
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Wei Yang
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Wendy Stam
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Alireza Mashaghi
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
26
|
Dawson HD, Chen C, Li RW, Bell LN, Shea-Donohue T, Kringel H, Beshah E, Hill DE, Urban JF. Molecular and metabolomic changes in the proximal colon of pigs infected with Trichuris suis. Sci Rep 2020; 10:12853. [PMID: 32732949 PMCID: PMC7393168 DOI: 10.1038/s41598-020-69462-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
The pig whipworm Trichuris suis is important in swine production because of its negative effects on pig performance and, notably, to some humans with inflammatory bowel disease as a therapeutic agent that modulates inflammation. The proximal colon of T. suis-infected pigs exhibited general inflammation around day 21 after inoculation with infective eggs that is transcriptionally characterized by markers of type-2 immune activation, inflammation, cellular infiltration, tissue repair enzymes, pathways of oxidative stress, and altered intestinal barrier function. Prominent gene pathways involved the Th2-response, de novo cholesterol synthesis, fructose and glucose metabolism, basic amino acid metabolism, and bile acid transport. Upstream regulatory factor analysis implicated the bile acid/farnesoid X receptor in some of these processes. Metabolic analysis indicated changes in fatty acids, antioxidant capacity, biochemicals related to methylation, protein glycosylation, extracellular matrix structure, sugars, Krebs cycle intermediates, microbe-derived metabolites and altered metabolite transport. Close to 1,200 differentially expressed genes were modulated in the proximal colon of pigs with a persistent adult worm infection that was nearly 90% lower in pigs that had expelled worms. The results support a model to test diets that favorably alter the microbiome and improve host intestinal health in both pigs and humans exposed to Trichuris.
Collapse
Affiliation(s)
- Harry D Dawson
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA
| | - Celine Chen
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA
| | - Robert W Li
- Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, USA
| | | | | | - Helene Kringel
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ethiopia Beshah
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA
| | - Dolores E Hill
- Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, USA
| | - Joseph F Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, USA. .,Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD, USA.
| |
Collapse
|
27
|
Xiong S, Hong Z, Huang LS, Tsukasaki Y, Nepal S, Di A, Zhong M, Wu W, Ye Z, Gao X, Rao GN, Mehta D, Rehman J, Malik AB. IL-1β suppression of VE-cadherin transcription underlies sepsis-induced inflammatory lung injury. J Clin Invest 2020; 130:3684-3698. [PMID: 32298238 PMCID: PMC7324198 DOI: 10.1172/jci136908] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022] Open
Abstract
Unchecked inflammation is a hallmark of inflammatory tissue injury in diseases such as acute respiratory distress syndrome (ARDS). Yet the mechanisms of inflammatory lung injury remain largely unknown. Here we showed that bacterial endotoxin lipopolysaccharide (LPS) and cecal ligation and puncture-induced (CLP-induced) polymicrobial sepsis decreased the expression of transcription factor cAMP response element binding (CREB) in lung endothelial cells. We demonstrated that endothelial CREB was crucial for VE-cadherin transcription and the formation of the normal restrictive endothelial adherens junctions. The inflammatory cytokine IL-1β reduced cAMP generation and CREB-mediated transcription of VE-cadherin. Furthermore, endothelial cell-specific deletion of CREB induced lung vascular injury whereas ectopic expression of CREB in the endothelium prevented the injury. We also observed that rolipram, which inhibits type 4 cyclic nucleotide phosphodiesterase-mediated (PDE4-mediated) hydrolysis of cAMP, prevented endotoxemia-induced lung vascular injury since it preserved CREB-mediated VE-cadherin expression. These data demonstrate the fundamental role of the endothelial cAMP-CREB axis in promoting lung vascular integrity and suppressing inflammatory injury. Therefore, strategies aimed at enhancing endothelial CREB-mediated VE-cadherin transcription are potentially useful in preventing sepsis-induced lung vascular injury in ARDS.
Collapse
Affiliation(s)
- Shiqin Xiong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Zhigang Hong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Long Shuang Huang
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Saroj Nepal
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Anke Di
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Ming Zhong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Wei Wu
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Zhiming Ye
- Department of Nephrology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaopei Gao
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Gadiparthi N. Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dolly Mehta
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
28
|
17β-estradiol regulates prostaglandin E 2 and F 2α synthesis and function in endometrial explants of cattle. Anim Reprod Sci 2020; 216:106466. [PMID: 32414468 DOI: 10.1016/j.anireprosci.2020.106466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 11/23/2022]
Abstract
Prostaglandins (PG) have primary functions in the reproductive tract, however, the mechanism of regulation of PG secretion in the endometrium is unclear. Estrogen as a predominant regulator of uterine functions during the mammalian estrous cycle and effects of estrogen on synthesis of PG and function in uterine tissues of cattle are not fully understood. In this study, there was evaluation of the concentration- and time-effects of 17β-estradiol on PG synthesis in endometrial explants of cattle, focusing on the secretion of prostaglandin E2 (PGE2) and prostaglandin F2α (PGF2α) as well as relative abundance of mRNA transcript and protein for both the enzymes responsible for PGE2 and PGF2α synthesis, including prostaglandin-endoperoxide synthase 1 and 2 (PTGS1, PTGS2), PGE2 synthase (PGES), PGF2α synthase (PGFS), and carbonyl reductase (CBR1), and the receptors responsible for downstream PGE2 (PTGER2, PTGER4) and PGF2α (PTGFR) signaling. Results indicated that 17β-estradiol increased PGE2 and PGF2α production at concentrations ranging from 10-11 to 10-8 M. Furthermore, abundances of PTGS1, PTGS2, PGES, PGFS, PTGER2, PTGER4, and PTGFR mRNA transcripts and protein were greater immediately after 17β-estradiol treatment at almost all the concentrations, while these CBR1 abundances were less as a result of treatments with 17β-estradiol. These data support the hypothesis that estradiol modulates the synthesis and function of PG in the endometrium of cattle.
Collapse
|
29
|
Hughes MJ, McGettrick HM, Sapey E. Shared mechanisms of multimorbidity in COPD, atherosclerosis and type-2 diabetes: the neutrophil as a potential inflammatory target. Eur Respir Rev 2020; 29:190102. [PMID: 32198215 PMCID: PMC9488696 DOI: 10.1183/16000617.0102-2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/02/2019] [Indexed: 12/17/2022] Open
Abstract
Multimorbidity is increasingly common and current healthcare strategies are not always aligned to treat this complex burden of disease. COPD, type-2 diabetes mellitus (T2D) and cardiovascular disease, especially atherosclerosis, occur more frequently together than expected, even when risk factors such as smoking, obesity, inactivity and poverty are considered. This supports the possibility of unifying mechanisms that contribute to the pathogenesis or progression of each condition.Neutrophilic inflammation is causally associated with COPD, and increasingly recognised in the pathogenesis of atherosclerosis and T2D, potentially forming an aetiological link between conditions. This link might reflect an overspill of inflammation from one affected organ into the systemic circulation, exposing all organs to an increased milieu of proinflammatory cytokines. Additionally, increasing evidence supports the involvement of other processes in chronic disease pathogenesis, such as cellular senescence or changes in cellular phenotypes.This review explores the current scientific evidence for inflammation, cellular ageing and cellular processes, such as reactive oxygen species production and phenotypic changes in the pathogenesis of COPD, T2D and atherosclerosis; highlighting common mechanisms shared across these diseases. We identify emerging therapeutic approaches that target these areas, but also where more work is still required to improve our understanding of the underlying cellular biology in a multimorbid disease setting.
Collapse
Affiliation(s)
- Michael J Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
30
|
Birukov KG, Oskolkova OV. The Good and Bad Faces of Oxidized Phospholipids: Friends or Foes of Vascular Endothelium? EUR J LIPID SCI TECH 2019. [DOI: 10.1002/ejlt.201800497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Konstantin G. Birukov
- Department of AnesthesiologyUMSOM Lung Biology ProgramUniversity of MarylandSchool of Medicine20 Penn Street, HSF‐2, Room S145Baltimore, MD21201USA
| | - Olga V. Oskolkova
- Institute of Pharmaceutical SciencesDepartment of Pharmaceutical ChemistryUniversity of Graz8020 GrazAustria
| |
Collapse
|
31
|
de Jesus Souza M, de Moraes JA, Da Silva VN, Helal-Neto E, Uberti AF, Scopel-Guerra A, Olivera-Severo D, Carlini CR, Barja-Fidalgo C. Helicobacter pylori urease induces pro-inflammatory effects and differentiation of human endothelial cells: Cellular and molecular mechanism. Helicobacter 2019; 24:e12573. [PMID: 30907046 DOI: 10.1111/hel.12573] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/21/2019] [Accepted: 01/30/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Helicobacter pylori urease (HPU) is a key virulence factor that enables bacteria to colonize and survive in the stomach. We early demonstrated that HPU, independent of its catalytic activity, induced inflammatory and angiogenic responses in vivo and directly activated human neutrophils to produce reactive oxygen species (ROS). We have investigated the effects of HPU on endothelial cells, focusing on the signaling mechanism involved. METHODS Monolayers of human microvascular endothelial cells (HMEC-1) were stimulated with HPU (up to 10 nmol/L): Paracellular permeability was accessed through dextran-FITC passage. NO and ROS production was evaluated using intracellular probes. Proteins or mRNA expressions were detected by Western blotting and fluorescence microscopy or qPCR assays, respectively. RESULTS Treatment with HPU enhanced paracellular permeability of HMEC-1, preceded by VE-cadherin phosphorylation and its dissociation from cell-cell junctions. This caused profound alterations in actin cytoskeleton dynamics and focal adhesion kinase (FAK) phosphorylation. HPU triggered ROS and nitric oxide (NO) production by endothelial cells. Increased intracellular ROS resulted in nuclear factor kappa B (NF-κB) activation and upregulated expression of cyclooxygenase-2 (COX-2), hemeoxygenase-1 (HO-1), interleukin-1β (IL-1β), and intercellular adhesion molecule-1 (ICAM-1). Higher ICAM-1 and E-selectin expression was associated with increased neutrophil adhesion on HPU-stimulated HMEC monolayers. The effects of HPU on endothelial cells were dependent on ROS production and lipoxygenase pathway activation, being inhibited by esculetin. Additionally, HPU improved vascular endothelial growth factor receptor 2 (VEGFR-2) expression. CONCLUSION The data suggest that the pro-inflammatory properties of HPU drive endothelial cell to a ROS-dependent program of differentiation that contributes to the progression of H pylori infection.
Collapse
Affiliation(s)
- Mariele de Jesus Souza
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Alfredo de Moraes
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Redox Biology, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vany Nascimento Da Silva
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edward Helal-Neto
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Augusto Frantz Uberti
- Laboratory of Neurotoxins, Brain Institute (BRAINS-InsCer), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Adriele Scopel-Guerra
- Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil
| | - Deiber Olivera-Severo
- Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil
| | - Célia R Carlini
- Laboratory of Neurotoxins, Brain Institute (BRAINS-InsCer), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil
| | - Christina Barja-Fidalgo
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Simmons S, Erfinanda L, Bartz C, Kuebler WM. Novel mechanisms regulating endothelial barrier function in the pulmonary microcirculation. J Physiol 2018; 597:997-1021. [PMID: 30015354 DOI: 10.1113/jp276245] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary epithelial and vascular endothelial cell layers provide two sequential physical and immunological barriers that together form a semi-permeable interface and prevent alveolar and interstitial oedema formation. In this review, we focus specifically on the continuous endothelium of the pulmonary microvascular bed that warrants strict control of the exchange of gases, fluid, solutes and circulating cells between the plasma and the interstitial space. The present review provides an overview of emerging molecular mechanisms that permit constant transcellular exchange between the vascular and interstitial compartment, and cause, prevent or reverse lung endothelial barrier failure under experimental conditions, yet with a clinical perspective. Based on recent findings and at times seemingly conflicting results we discuss emerging paradigms of permeability regulation by altered ion transport as well as shifts in the homeostasis of sphingolipids, angiopoietins and prostaglandins.
Collapse
Affiliation(s)
- Szandor Simmons
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lasti Erfinanda
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bartz
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Cortactin: Cell Functions of A Multifaceted Actin-Binding Protein. Trends Cell Biol 2018; 28:79-98. [DOI: 10.1016/j.tcb.2017.10.009] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022]
|
34
|
Abstract
Lipid mediators play a critical role in the development and resolution of vascular endothelial barrier dysfunction caused by various pathologic interventions. The accumulation of excess lipids directly impairs endothelial cell (EC) barrier function that is known to contribute to the development of atherosclerosis and metabolic disorders such as obesity and diabetes as well as chronic inflammation in the vascular endothelium. Certain products of phospholipid oxidation (OxPL) such as fragmented phospholipids generated during oxidative and nitrosative stress show pro-inflammatory potential and cause endothelial barrier dysfunction. In turn, other OxPL products enhance basal EC barrier and exhibit potent barrier-protective effects in pathologic settings of acute vascular leak caused by pro-inflammatory mediators, barrier disruptive agonists and pathologic mechanical stimulation. These beneficial effects were further confirmed in rodent models of lung injury and inflammation. The bioactive oxidized lipid molecules may serve as important therapeutic prototype molecules for future treatment of acute lung injury syndromes associated with endothelial barrier dysfunction and inflammation. This review will summarize recent studies of biological effects exhibited by various groups of lipid mediators with a focus on the role of oxidized phospholipids in control of vascular endothelial barrier, agonist induced EC permeability, inflammation, and barrier recovery related to clinical settings of acute lung injury and inflammatory vascular leak.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland Baltimore, School of Medicine, Baltimore, MD, USA
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland Baltimore, School of Medicine, Baltimore, MD, USA,CONTACT Konstantin G. Birukov, MD, PhD Department of Anesthesiology, University of Maryland, School of Medicine, 20 Penn Street, HSF-2, Room 145, Baltimore, MD 21201, USA
| |
Collapse
|