1
|
Chaudhary N, Kasiewicz LN, Newby AN, Arral ML, Yerneni SS, Melamed JR, LoPresti ST, Fein KC, Strelkova Petersen DM, Kumar S, Purwar R, Whitehead KA. Amine headgroups in ionizable lipids drive immune responses to lipid nanoparticles by binding to the receptors TLR4 and CD1d. Nat Biomed Eng 2024:10.1038/s41551-024-01256-w. [PMID: 39363106 DOI: 10.1038/s41551-024-01256-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 09/05/2024] [Indexed: 10/05/2024]
Abstract
Lipid nanoparticles (LNPs) are the most clinically advanced delivery vehicle for RNA therapeutics, partly because of established lipid structure-activity relationships focused on formulation potency. Yet such knowledge has not extended to LNP immunogenicity. Here we show that the innate and adaptive immune responses elicited by LNPs are linked to their ionizable lipid chemistry. Specifically, we show that the amine headgroups in ionizable lipids drive LNP immunogenicity by binding to Toll-like receptor 4 and CD1d and by promoting lipid-raft formation. Immunogenic LNPs favour a type-1 T-helper-cell-biased immune response marked by increases in the immunoglobulins IgG2c and IgG1 and in the pro-inflammatory cytokines tumour necrosis factor, interferon γ and the interleukins IL-6 and IL-2. Notably, the inflammatory signals originating from these receptors inhibit the production of anti-poly(ethylene glycol) IgM antibodies, preventing the often-observed loss of efficacy in the LNP-mediated delivery of siRNA and mRNA. Moreover, we identified computational methods for the prediction of the structure-dependent innate and adaptive responses of LNPs. Our findings may help accelerate the discovery of well-tolerated ionizable lipids suitable for repeated dosing.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Lisa N Kasiewicz
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alexandra N Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Mariah L Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Jilian R Melamed
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Samuel T LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Katherine C Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Sushant Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Rahul Purwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Chen J, Jiang M, Ying Y, Ji Y, Chi Y, Tao L, Wu F, Chen M. Network pharmacological mechanism analysis and evidence-based medical validation of Dahuang Mudan Decoction in the treatment of acute pancreatitis. Medicine (Baltimore) 2024; 103:e39679. [PMID: 39287237 PMCID: PMC11404899 DOI: 10.1097/md.0000000000039679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Dahuang Mudan Decoction is commonly used in China for the treatment of acute pancreatitis. Nevertheless, the therapeutic efficacy of the drug remains a subject of debate, and its active ingredients and potential therapeutic targets remain to be determined. The present study used a network pharmacological approach to investigate the active ingredients and possible targets of the drug, and illustrated the clinical effectiveness of Dahuang Mudan Decoction in the treatment of acute pancreatitis by meta-analysis. METHODS The present study investigated the active ingredients of the constituent herbs of Dahuang Mudan Decoction using the TCMID database. In order to further identify molecular targets, Swiss Target Prediction, OMIM and Genecards databases was be used. The present study used metascape database for gene ontology function enrichment analysis and Kyoto Genome Encyclopedia pathway enrichment analysis. A gene interaction network diagram was established for predicting the main targets and mechanism of action to Dahuang Mudan Decoction for acute pancreatitis. To further illustrate the validity of the gene targets and the clinical efficacy of the drug, 13 relevant studies were included for meta-analysis and analyzed using the Cochrane Collaboration's Review Manager 5.4 software. RESULT After a thorough screening process, the present study identified three main components of Dahuang Mudan Decoction: kaempferol, quercetin and eupatin. These three major components have the potential to target 5 important proteins: AKT1, TNF-a, IL-6, TP53, HIF1A. In addition, pathway analyses by the Kyoto Genome Encyclopedia showed that Dahuang Mudan Decoction is active through the Pathways in cancer, AGE-RAGE signaling pathway in diabetic complications, PI3K-Akt signaling pathway, etc signaling pathway to act on acute pancreatitis. The results of meta-analysis showed that compared with the control group, the experimental group had superior performance in terms of overall treatment efficacy, reduction of hospital stays and inflammatory factor levels after treatment. CONCLUSION In summary, network pharmacological studies have shown that Dahuang Mudan Decoction affects acute pancreatitis through different components, targets, and mechanisms. In addition, the meta-analysis study strongly supported the effectiveness of Dahuang Mudan Decoction in the treatment of acute pancreatitis.
Collapse
Affiliation(s)
- Jinhan Chen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengjie Jiang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuou Ying
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Ji
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuying Chi
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Linghui Tao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fuping Wu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Mingxian Chen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
3
|
Sawoo R, Bishayi B. TLR4/TNFR1 blockade suppresses STAT1/STAT3 expression and increases SOCS3 expression in modulation of LPS-induced macrophage responses. Immunobiology 2024; 229:152840. [PMID: 39126792 DOI: 10.1016/j.imbio.2024.152840] [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: 12/15/2023] [Revised: 07/15/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024]
Abstract
Due to the urgent need to create appropriate treatment techniques, which are currently unavailable, LPS-induced sepsis has become a serious concern on a global scale. The primary active component in the pathophysiology of inflammatory diseases such as sepsis is the Gram-negative bacterial lipopolysaccharide (LPS). LPS interacts with cell surface TLR4 in macrophages, causing the formation of reactive oxygen species (ROS), TNF-α, IL-1β and oxidative stress. It also significantly activates the MAPKs and NF-κB pathway. Excessive production of pro-inflammatory cytokines is one of the primary characteristic features in the onset and progression of inflammation. Cytokines mainly signal through the JAK/STAT pathway. We hypothesize that blocking of TLR4 along with TNFR1 might be beneficial in suppressing the effects of STAT1/STAT3 due to the stimulation of SOCS3 proteins. Prior to the LPS challenge, the macrophages were treated with antibodies against TLR4 and TNFR1 either individually or in combination. On analysis of the macrophage populations by flowcytometry, it was seen that receptor blockade facilitated the phenotypic shift of the M1 macrophages towards M2 resulting in lowered oxidative stress. Blocking of TLR4/TNFR1 upregulated the SOCS3 and mTOR expressions that enabled the transition of inflammatory M1 macrophages towards the anti-inflammatory M2 phenotype, which might be crucial in curbing the inflammatory responses. Also the reduction in the production of inflammatory cytokines such as IL-6, IL-1β due to the reduction in the activation of the STAT1 and STAT3 molecules was observed in our combination treatment group. All these results indicated that neutralization of both TLR4 and TNFR1 might provide new insights in establishing an alternative therapeutic strategy for LPS-sepsis.
Collapse
Affiliation(s)
- Ritasha Sawoo
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| |
Collapse
|
4
|
Abdel-Aziz N, Saif-Elnasr M. Citicoline modulates inflammatory signaling pathways in the spleen of rats exposed to gamma-radiation. Immunopharmacol Immunotoxicol 2024:1-8. [PMID: 39049671 DOI: 10.1080/08923973.2024.2381759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND AND AIM The spleen has an essential role in immune responses regulation and is considered the biggest peripheral immune organ. Citicoline is used for various brain disorders management. This study aimed to examine the using possibility of citicoline to treat γ-radiation-induced splenic inflammation in rats. MATERIALS AND METHODS Eighteen male albino rats were classified into: Group 1 (control) animals were kept as control. Group 2 (γ-radiation) animals were total-body γ-irradiated with 6 Gy. Group 3 (γ-radiation + citicoline) rats were γ-irradiated with 6 Gy, then injected intraperitoneally with citicoline (300 mg/kg/d) 5 min after irradiation for one week. Levels of TNF-α, IL-1β, iNOS, NF-κB, JAK2, and STAT3 were determined in spleen tissue, along with histopathological examination. RESULTS Rats exposure to gamma-radiation led to elevation in splenic TNF-α, IL-1β, NF-κB, iNOS, JAK2, and STAT3 levels significantly. Treatment with citicoline after gamma-radiation exposure improved this elevation, and modulated gamma-radiation-induced histopathological alterations. CONCLUSIONS This data showed that citicoline inhibited γ-radiation-induced splenic inflammation via suppressing NF-κB and JAK2/STAT3 signaling pathways in spleen tissue.
Collapse
Affiliation(s)
- Nahed Abdel-Aziz
- Radiation Biology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mostafa Saif-Elnasr
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
5
|
Zhou S, Sun Q, Gao N, Hu Z, Jia J, Song J, Xu G, Dong A, Xia W, Wu J. The Role of Inflammatory Biomarkers in Mediating the Effect of Inflammatory Bowel Disease on nonmalignant Digestive System Diseases: A Multivariable Mendelian Randomized Study. Can J Gastroenterol Hepatol 2024; 2024:1266139. [PMID: 38529201 PMCID: PMC10963109 DOI: 10.1155/2024/1266139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/03/2024] [Accepted: 03/10/2024] [Indexed: 03/27/2024] Open
Abstract
Background While observation studies have shown a positive correlation between inflammatory bowel disease (IBD) and the risk of nonmalignant digestive system diseases, a definitive causal relationship has not yet been clearly established. Methods Mendelian randomization (MR) was employed to investigate the potential causal association between genetic susceptibility to IBD and nonmalignant gastrointestinal diseases. Genetic variants were extracted as instrumental variables (IVs) from a genome-wide association study (GWAS) meta-analysis, which included 12,194 cases of Crohn's disease (CD) and 28,072 control cases of European ancestry. The GWAS for ulcerative colitis (UC) included 12,366 UC and 33,609 control cases of European ancestry. All IVs reached genome-wide significance (GWAS p value <5 × 10-8). Summary-level data for acute pancreatitis (AP), irritable bowel syndrome (IBS), gastroesophageal reflux disease, cholelithiasis, and CeD (celiac disease) were obtained from the GWAS meta-analysis and the FinnGen dataset. Summary-level data on relevant inflammatory factors were provided by the International Genetic Consortium. Univariate MR analysis was conducted using inverse variance weighting as the primary method for estimating causal effects. Multivariate MR analyses were also performed to detect possible mediators. Results Genetic susceptibility to UC was associated with an increased risk of AP (OR = 1.08; 95% CI = 1.03-1.13; p=0.002) and IBS odds ratio (OR] = 1.07; 95% confidence interval (CI] = 1.03-1.11; (p < 0.001). In terms of potential mediators, interleukin 6 (IL-6) had a driving effect on the association between UC and AP. There was no apparent evidence of increased risk with CD. Meanwhile, genetic susceptibility to CD increases the risk of CeD (OR = 1.14; 95% CI = 1.03-1.25; p=0.01). Conclusions The evidence suggests that UC is associated with an elevated risk of AP and IBS, and IL-6 may be responsible in AP. CD is associated with an increased risk of developing CeD. Implementing a proactive monitoring program for assessing the risk of gastrointestinal diseases in UC patients, particularly those with elevated IL-6 levels, may be of interest. In addition, the presence of AP and IBS may indicate the presence of UC. Preventing CeD is an essential consideration in the therapeutic management of patients with CD.
Collapse
Affiliation(s)
- Shu Zhou
- Hangzhou Ninth People's Hospital, Hangzhou, China
| | - Qi Sun
- Hangzhou Ninth People's Hospital, Hangzhou, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
| | - Ning Gao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zekai Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
| | - Junjun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
| | | | - Guocong Xu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Aiqiang Dong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiliang Xia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
| | - Jiafeng Wu
- Hangzhou Ninth People's Hospital, Hangzhou, China
| |
Collapse
|
6
|
Silva AR, de Souza e Souza KFC, Souza TBD, Younes-Ibrahim M, Burth P, de Castro Faria Neto HC, Gonçalves-de-Albuquerque CF. The Na/K-ATPase role as a signal transducer in lung inflammation. Front Immunol 2024; 14:1287512. [PMID: 38299144 PMCID: PMC10827986 DOI: 10.3389/fimmu.2023.1287512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is marked by damage to the capillary endothelium and alveolar epithelium following edema formation and cell infiltration. Currently, there are no effective treatments for severe ARDS. Pathologies such as sepsis, pneumonia, fat embolism, and severe trauma may cause ARDS with respiratory failure. The primary mechanism of edema clearance is the epithelial cells' Na/K-ATPase (NKA) activity. NKA is an enzyme that maintains the electrochemical gradient and cell homeostasis by transporting Na+ and K+ ions across the cell membrane. Direct injury on alveolar cells or changes in ion transport caused by infections decreases the NKA activity, loosening tight junctions in epithelial cells and causing edema formation. In addition, NKA acts as a receptor triggering signal transduction in response to the binding of cardiac glycosides. The ouabain (a cardiac glycoside) and oleic acid induce lung injury by targeting NKA. Besides enzymatic inhibition, the NKA triggers intracellular signal transduction, fostering proinflammatory cytokines production and contributing to lung injury. Herein, we reviewed and discussed the crucial role of NKA in edema clearance, lung injury, and intracellular signaling pathway activation leading to lung inflammation, thus putting the NKA as a protagonist in lung injury pathology.
Collapse
Affiliation(s)
- Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Thamires Bandeira De Souza
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Mauricio Younes-Ibrahim
- Departamento de Medicina Interna, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Gál R, Halmosi R, Gallyas F, Tschida M, Mutirangura P, Tóth K, Alexy T, Czopf L. Resveratrol and beyond: The Effect of Natural Polyphenols on the Cardiovascular System: A Narrative Review. Biomedicines 2023; 11:2888. [PMID: 38001889 PMCID: PMC10669290 DOI: 10.3390/biomedicines11112888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases (CVDs) are among the leading causes of morbidity and mortality worldwide. Unhealthy dietary habits have clearly been shown to contribute to the development of CVDs. Beyond the primary nutrients, a healthy diet is also rich in plant-derived compounds. Natural polyphenols, found in fruits, vegetables, and red wine, have a clear role in improving cardiovascular health. In this review, we strive to summarize the results of the relevant pre-clinical and clinical trials that focused on some of the most important natural polyphenols, such as resveratrol and relevant flavonoids. In addition, we aim to identify their common sources, biosynthesis, and describe their mechanism of action including their regulatory effect on signal transduction pathways. Finally, we provide scientific evidence regarding the cardiovascular benefits of moderate, long-term red wine consumption.
Collapse
Affiliation(s)
- Roland Gál
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary; (R.G.); (R.H.); (K.T.)
- Szentágothai Research Centre, University of Pecs, 7624 Pecs, Hungary
| | - Róbert Halmosi
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary; (R.G.); (R.H.); (K.T.)
- Szentágothai Research Centre, University of Pecs, 7624 Pecs, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pecs, 7624 Pecs, Hungary;
| | - Michael Tschida
- Medical School, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Pornthira Mutirangura
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Kálmán Tóth
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary; (R.G.); (R.H.); (K.T.)
- Szentágothai Research Centre, University of Pecs, 7624 Pecs, Hungary
| | - Tamás Alexy
- Department of Medicine, Division of Cardiology, University of Minnesota, Minneapolis, MN 55455, USA;
| | - László Czopf
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary; (R.G.); (R.H.); (K.T.)
| |
Collapse
|
8
|
Zerikiotis S, Efentakis P, Dapola D, Agapaki A, Seiradakis G, Kostomitsopoulos N, Skaltsounis AL, Tseti I, Triposkiadis F, Andreadou I. Synergistic Pulmonoprotective Effect of Natural Prolyl Oligopeptidase Inhibitors in In Vitro and In Vivo Models of Acute Respiratory Distress Syndrome. Int J Mol Sci 2023; 24:14235. [PMID: 37762537 PMCID: PMC10531912 DOI: 10.3390/ijms241814235] [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: 08/23/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a highly morbid inflammatory lung disease with limited pharmacological interventions. The present study aims to evaluate and compare the potential pulmonoprotective effects of natural prolyl oligopeptidase (POP) inhibitors namely rosmarinic acid (RA), chicoric acid (CA), epigallocatechin-3-gallate (EGCG) and gallic acid (GA), against lipopolysaccharide (LPS)-induced ARDS. Cell viability and expression of pro-inflammatory mediators were measured in RAW264.7 cells and in primary murine lung epithelial and bone marrow cells. Nitric oxide (NO) production was also assessed in unstimulated and LPS-stimulated RAW264.7 cells. For subsequent in vivo experiments, the two natural products (NPs) with the most favorable effects, RA and GA, were selected. Protein, cell content and lipid peroxidation levels in bronchoalveolar lavage fluid (BALF), as well as histopathological changes and respiratory parameters were evaluated in LPS-challenged mice. Expression of key mediators involved in ARDS pathophysiology was detected by Western blotting. RA and GA favorably reduced gene expression of pro-inflammatory mediators in vitro, while GA decreased NO production in macrophages. In LPS-challenged mice, RA and GA co-administration improved respiratory parameters, reduced cell and protein content and malondialdehyde (MDA) levels in BALF, decreased vascular cell adhesion molecule-1 (VCAM-1) and the inducible nitric oxide synthase (iNOS) protein expression, activated anti-apoptotic mechanisms and down-regulated POP in the lung. Conclusively, these synergistic pulmonoprotective effects of RA and GA co-administration could render them a promising prophylactic/therapeutic pharmacological intervention against ARDS.
Collapse
Affiliation(s)
- Stelios Zerikiotis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece; (S.Z.); (P.E.); (D.D.); (G.S.)
| | - Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece; (S.Z.); (P.E.); (D.D.); (G.S.)
| | - Danai Dapola
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece; (S.Z.); (P.E.); (D.D.); (G.S.)
| | - Anna Agapaki
- Histochemistry Facility, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece;
| | - Georgios Seiradakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece; (S.Z.); (P.E.); (D.D.); (G.S.)
| | - Nikolaos Kostomitsopoulos
- Laboratory Animal Facility, Centre of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 115 27 Athens, Greece;
| | - Alexios-Leandros Skaltsounis
- Section of Pharmacognosy and Natural Product Chemistry Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece;
| | | | - Filippos Triposkiadis
- Department of Cardiology, University General Hospital of Larissa, 413 34 Larissa, Greece;
- Faculty of Health Sciences, University of Thessaly, 413 34 Larissa, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, 157 71 Athens, Greece; (S.Z.); (P.E.); (D.D.); (G.S.)
| |
Collapse
|
9
|
Deng W, Lu Y, Hu P, Zhang Q, Li S, Yang D, Zhao N, Qian K, Liu F. Integrated Analysis of Non-Coding RNA and mRNA Expression Profiles in Exosomes from Lung Tissue with Sepsis-Induced Acute Lung Injury. J Inflamm Res 2023; 16:3879-3895. [PMID: 37674532 PMCID: PMC10478974 DOI: 10.2147/jir.s419491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/19/2023] [Indexed: 09/08/2023] Open
Abstract
Background Acute lung injury (ALI) is associated with a high mortality rate; however, the underlying molecular mechanisms are poorly understood. The purpose of this study was to investigate the expression profile and related networks of long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and mRNAs in lung tissue exosomes obtained from sepsis-induced ALI. Methods A mouse model of sepsis was established using the cecal ligation and puncture method. RNA sequencing was performed using lung tissue exosomes obtained from mice in the sham and CLP groups. Hematoxylin-eosin staining, Western blotting, immunofluorescence, quantitative real-time polymerase chain reaction, and nanoparticle tracking analysis were performed to identify relevant phenotypes, and bioinformatic algorithms were used to evaluate competitive endogenous RNA (ceRNA) networks. Results Thirty lncRNA-miRNA-mRNA interactions were identified, including two upregulated lncRNAs, 30 upregulated miRNAs, and two downregulated miRNAs. Based on the expression levels of differentially expressed mRNAs(DEmRNAs), differentially expressed LncRNAs(DELncRNAs), and differentially expressed miRNAs(DEmiRNAs), 30 ceRNA networks were constructed. Conclusion Our study revealed, for the first time, the expression profiles of lncRNA, miRNA, and mRNA in exosomes isolated from the lungs of mice with sepsis-induced ALI, and the exosome co-expression network and ceRNA network related to ALI in sepsis.
Collapse
Affiliation(s)
- Wei Deng
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Yanhua Lu
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Ping Hu
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Qingqing Zhang
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Shuangyan Li
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Dong Yang
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Ning Zhao
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Kejian Qian
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Fen Liu
- Department of Critical Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
- Medical Innovation Center, First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
10
|
Li G, Ma J, Yang Y, Zang C, Ju C, Yuan F, Ning J, Shang M, Chen Q, Jiang Y, Li F, Bao X, Mu D, Zhang D. Yinma Jiedu Granule attenuates LPS-induced acute lung injury in rats via suppressing inflammation level. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116292. [PMID: 36931412 DOI: 10.1016/j.jep.2023.116292] [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: 12/04/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yinma Jiedu Granule (YMJD) is a traditional Chinese patent medicine (CPM), which has been proved to have anti-inflammatory effects and therapeutical effects on obstructive pulmonary disease. AIM OF STUDY The purpose of the current investigation is to find out if YMJD can alleviate acute lung injury (ALI) induced by lipopolysaccharide (LPS) in rats and its underlying mechanisms. MATERIALS AND METHODS Rats were treated with either vehicle or YMJD for 14 consecutive days, and 2 h after the last administration, the rat model of ALI was induced by the intratracheal instillation of LPS. High performance liquid chromatography (HPLC) was applied for the fingerprint analysis of YMJD. The efficacy and molecular mechanisms were investigated. RESULTS The results showed that treatment with YMJD improved the general state of rats, reduced weight loss and serum lactate (LA) levels, attenuated pulmonary edema and pathological damage of the lung tissue. Moreover, we found that YMJD effectively decreased the infiltration of white blood cells (WBC), lymphocytes (LYM), mononuclear cells (MON) and neutrophils (NEUT) in bronchoalveolar lavage fluid (BALF), reduced the concentration of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) and inhibited inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression in the lung tissue. Additionally, we found that YMJD could significantly increase the activity of superoxide dismutase (SOD) and reduce the malondialdehyde (MDA) level in the lung tissue. By employing RNA-sequencing, we have identified that JAK2/STAT1 is an important pathway that is involved in the lung protection of YMJD, and further Western blot assay verified that YMJD could effectively inhibit the activation of the JAK2/STAT1 pathway. CONCLUSIONS YMJD could attenuate LPS-induced ALI through suppressing inflammation and oxidative stress in the lung tissue of rats, associating with the inhibition of JAK2/STAT1 activation. These findings provide evidence for the clinical use of YMJD for treatment of inflammatory pulmonary diseases like ALI.
Collapse
Affiliation(s)
- Gen Li
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jingwei Ma
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yang Yang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Caixia Zang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Cheng Ju
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Fangyu Yuan
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jingwen Ning
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Meiyu Shang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qiuzhu Chen
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yueqi Jiang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Fangfang Li
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiuqi Bao
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Degui Mu
- Fudan University, Shanghai, China.
| | - Dan Zhang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Zhang K, Li C, Sun J, Tian X. PRMT5 inhibition ameliorates inflammation and promotes the osteogenic differentiation of LPS‑induced periodontal stem cells via STAT3/NF‑κB signaling. Exp Ther Med 2023; 25:264. [PMID: 37206565 PMCID: PMC10189754 DOI: 10.3892/etm.2023.11963] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/06/2022] [Indexed: 05/21/2023] Open
Abstract
It has been reported that protein arginine methyltransferase 5 (PRMT5) serves a significant role in osteogenic differentiation and inflammatory response. Nevertheless, its role in periodontitis as well as its underlying mechanism remain to be elucidated. The aim of the present study was to explore the role of PRMT5 in periodontitis and whether PRMT5 could reduce liposaccharide (LPS)-induced inflammation of human periodontal ligament stem cells (hPDLSCs) and promote osteogenic differentiation through STAT3/NF-κB signaling. In the current study, the expression levels of PRMT5 were determined in LPS-induced hPDLSCs by reverse transcription-quantitative PCR and western blot analysis. ELISA and western blot analysis were employed to assess the secretion and expression levels of inflammatory factors, respectively. The osteogenic differentiation and mineralization potential of hPDLSCs were evaluated using alkaline phosphatase (ALP) activity assay, Alizarin red staining and western blot analysis. Additionally, western blot analysis was applied to determine the expression levels of the STAT3/NF-κB signaling pathway-related proteins. The results showed that the expression levels of PRMT5 were significantly enhanced in LPS-induced hPDLSCs. Additionally, PRMT5 knockdown reduced the contents of IL-1β, IL-6, TNF-α, inducible nitric oxide synthase and cyclooxygenase-2. PRMT5 depletion also enhanced ALP activity, improved the mineralization ability and upregulated bone morphogenetic protein 2, osteocalcin and runt-related transcription factor 2 in LPS-induced hPDLSCs. Furthermore, PRMT5 knockdown inhibited inflammation and promoted the osteogenic differentiation of hPDLSCs via blocking the activation of the STAT3/NF-κB signaling pathway. In conclusion, PRMT5 inhibition suppressed LPS-induced inflammation and accelerated osteogenic differentiation in hPDLSCs via regulating STAT3/NF-κB signaling, thus providing a potential targeted therapy for the improvement of periodontitis.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Changshun Li
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jian Sun
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Xiaobei Tian
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- Correspondence to: Professor Xiaobei Tian, Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, 130 Huaihai West Road, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
12
|
Zhao M, Wang X, Kumar SA, Yao Y, Sun M. A Pharmacological Insight of Piperlongumine, Bioactive Validating Its Therapeutic Efficacy as a Drug to Treat Inflammatory Diseases. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2023. [DOI: 10.1134/s1068162023020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
13
|
Zhu H, Zhang H, Lu K, Yang S, Tang X, Zhou M, Sun G, Zhang Z, Chu H. Chlorinated Organophosphate Flame Retardants Impair the Lung Function via the IL-6/JAK/STAT Signaling Pathway. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:17858-17869. [PMID: 36480654 DOI: 10.1021/acs.est.2c05357] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Toxicological studies have revealed the adverse impacts of organophosphate flame retardants (OPFRs) on the respiratory system, while there is a lack of epidemiological evidence, and information for risk assessment remains insufficient. Herein, we investigated the associations of urinary metabolites of OPFRs with the lung function in 987 adults participating in the U.S. National Health and Nutrition Examination Survey 2011-2012. The elevation of three primary metabolites of chlorinated OPFRs [bis(1,3-dichloro-2-propyl) phosphate (BDCIPP), bis(2-chloroethyl) phosphate (BCEP), and bis(1-chloro-2-propyl) phosphate (BCIPP)] was related to pulmonary dysfunction in a sample-weighted regression model. Each one-unit increase in the log-transformed levels of BDCIPP and BCEP was related to 91.52 and 79.34 mL reductions in the forced vital capacity (FVC). Each one-unit elevation in BCIPP was correlated with 130.86, 153.56, 302.26, and 148.24 mL reductions in forced expiratory volume 1st second (FEV1), FVC, peak expiratory flow rate (PEF), and forced expiratory flow at 25-75% of FVC (FEF25-75%), respectively. Then, an adverse outcome pathway (AOP) framework was constructed using the Comparative Toxicogenomics Database, the Toxicity Forecaster, and the GeneCards database. Based on the weight of the evidence, BDCIPP, BCEP, BCIPP, and their parent compounds (TDCIPP, TCEP, and TCIPP) may affect the IL-6/Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway, induce airway remodeling, and impair the lung function. Additionally, tobacco smoke exposure may modify the effects of BDCIPP on the lung function (Pint < 0.05) and affect the IL-6-mediated AOP. These results suggested that chlorinated OPFRs were associated with pulmonary dysfunction via the IL-6/JAK/STAT pathway.
Collapse
Affiliation(s)
- Huanhuan Zhu
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Huilin Zhang
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Kai Lu
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Sheng Yang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xiying Tang
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Meiyu Zhou
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Guanting Sun
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Haiyan Chu
- Department of Environmental Genomics, Institute of Healthy Jiangsu Development, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| |
Collapse
|
14
|
Batra R, Whalen W, Alvarez-Mulett S, Gomez-Escobar LG, Hoffman KL, Simmons W, Harrington J, Chetnik K, Buyukozkan M, Benedetti E, Choi ME, Suhre K, Schenck E, Choi AMK, Schmidt F, Cho SJ, Krumsiek J. Multi-omic comparative analysis of COVID-19 and bacterial sepsis-induced ARDS. PLoS Pathog 2022; 18:e1010819. [PMID: 36121875 PMCID: PMC9484674 DOI: 10.1371/journal.ppat.1010819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/19/2022] [Indexed: 12/06/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS), a life-threatening condition characterized by hypoxemia and poor lung compliance, is associated with high mortality. ARDS induced by COVID-19 has similar clinical presentations and pathological manifestations as non-COVID-19 ARDS. However, COVID-19 ARDS is associated with a more protracted inflammatory respiratory failure compared to traditional ARDS. Therefore, a comprehensive molecular comparison of ARDS of different etiologies groups may pave the way for more specific clinical interventions. METHODS AND FINDINGS In this study, we compared COVID-19 ARDS (n = 43) and bacterial sepsis-induced (non-COVID-19) ARDS (n = 24) using multi-omic plasma profiles covering 663 metabolites, 1,051 lipids, and 266 proteins. To address both between- and within- ARDS group variabilities we followed two approaches. First, we identified 706 molecules differently abundant between the two ARDS etiologies, revealing more than 40 biological processes differently regulated between the two groups. From these processes, we assembled a cascade of therapeutically relevant pathways downstream of sphingosine metabolism. The analysis suggests a possible overactivation of arginine metabolism involved in long-term sequelae of ARDS and highlights the potential of JAK inhibitors to improve outcomes in bacterial sepsis-induced ARDS. The second part of our study involved the comparison of the two ARDS groups with respect to clinical manifestations. Using a data-driven multi-omic network, we identified signatures of acute kidney injury (AKI) and thrombocytosis within each ARDS group. The AKI-associated network implicated mitochondrial dysregulation which might lead to post-ARDS renal-sequalae. The thrombocytosis-associated network hinted at a synergy between prothrombotic processes, namely IL-17, MAPK, TNF signaling pathways, and cell adhesion molecules. Thus, we speculate that combination therapy targeting two or more of these processes may ameliorate thrombocytosis-mediated hypercoagulation. CONCLUSION We present a first comprehensive molecular characterization of differences between two ARDS etiologies-COVID-19 and bacterial sepsis. Further investigation into the identified pathways will lead to a better understanding of the pathophysiological processes, potentially enabling novel therapeutic interventions.
Collapse
Affiliation(s)
- Richa Batra
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - William Whalen
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Sergio Alvarez-Mulett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Luis G. Gomez-Escobar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Katherine L. Hoffman
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, New York, United States of America
| | - Will Simmons
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, New York, United States of America
| | - John Harrington
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Kelsey Chetnik
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Mustafa Buyukozkan
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Elisa Benedetti
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Mary E. Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, New York, United States of America
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine–Qatar, Qatar Foundation, Doha, Qatar
| | - Edward Schenck
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Augustine M. K. Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine–Qatar, Qatar Foundation, Doha, Qatar
| | - Soo Jung Cho
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| |
Collapse
|
15
|
Batra R, Whalen W, Alvarez-Mulett S, Gómez-Escobar LG, Hoffman KL, Simmons W, Harrington J, Chetnik K, Buyukozkan M, Benedetti E, Choi ME, Suhre K, Schenck E, Choi AMK, Schmidt F, Cho SJ, Krumsiek J. Multi-omic comparative analysis of COVID-19 and bacterial sepsis-induced ARDS. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.05.16.22274587. [PMID: 35982655 PMCID: PMC9387161 DOI: 10.1101/2022.05.16.22274587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Acute respiratory distress syndrome (ARDS), a life-threatening condition characterized by hypoxemia and poor lung compliance, is associated with high mortality. ARDS induced by COVID-19 has similar clinical presentations and pathological manifestations as non-COVID-19 ARDS. However, COVID-19 ARDS is associated with a more protracted inflammatory respiratory failure compared to traditional ARDS. Therefore, a comprehensive molecular comparison of ARDS of different etiologies groups may pave the way for more specific clinical interventions. Methods and Findings In this study, we compared COVID-19 ARDS (n=43) and bacterial sepsis-induced (non-COVID-19) ARDS (n=24) using multi-omic plasma profiles covering 663 metabolites, 1,051 lipids, and 266 proteins. To address both between- and within-ARDS group variabilities we followed two approaches. First, we identified 706 molecules differently abundant between the two ARDS etiologies, revealing more than 40 biological processes differently regulated between the two groups. From these processes, we assembled a cascade of therapeutically relevant pathways downstream of sphingosine metabolism. The analysis suggests a possible overactivation of arginine metabolism involved in long-term sequelae of ARDS and highlights the potential of JAK inhibitors to improve outcomes in bacterial sepsis-induced ARDS. The second part of our study involved the comparison of the two ARDS groups with respect to clinical manifestations. Using a data-driven multi-omic network, we identified signatures of acute kidney injury (AKI) and thrombocytosis within each ARDS group. The AKI-associated network implicated mitochondrial dysregulation which might lead to post-ARDS renal-sequalae. The thrombocytosis-associated network hinted at a synergy between prothrombotic processes, namely IL-17, MAPK, TNF signaling pathways, and cell adhesion molecules. Thus, we speculate that combination therapy targeting two or more of these processes may ameliorate thrombocytosis-mediated hypercoagulation. Conclusion We present a first comprehensive molecular characterization of differences between two ARDS etiologies - COVID-19 and bacterial sepsis. Further investigation into the identified pathways will lead to a better understanding of the pathophysiological processes, potentially enabling novel therapeutic interventions.
Collapse
Affiliation(s)
- Richa Batra
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - William Whalen
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sergio Alvarez-Mulett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Luis G Gómez-Escobar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Katherine L Hoffman
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, NY, USA
| | - Will Simmons
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, NY, USA
| | - John Harrington
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kelsey Chetnik
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mustafa Buyukozkan
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Elisa Benedetti
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine - Qatar, Qatar Foundation, Doha, Qatar
| | - Edward Schenck
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Augustine M K Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine - Qatar, Qatar Foundation, Doha, Qatar
| | - Soo Jung Cho
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
16
|
Shen Z, Sun F, Lu Y, Yuan L, Ge S, Gong Q, Shi H. High temperature requirement A3 attenuates hypoxia/reoxygenation induced injury in H9C2 cells via suppressing inflammatory responses. Eur J Pharmacol 2022; 928:175114. [PMID: 35764130 DOI: 10.1016/j.ejphar.2022.175114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022]
Abstract
High temperature requirement A3 (HtrA3) belongs to the HtrA family, and its role in inflammation and myocardial ischemia-reperfusion injury remains unknown. Herein, the study aimed to explore the role of HtrA3 in inflammatory cytokine secretion and the nuclear factor kappa B (NF-κB) signaling pathway in hypoxia-reoxygenation (H/R)-induced H9C2 cardiomyoblasts. H9C2 cells were treated with H/R to mimic myocardial ischemia-reperfusion in vitro. Results showed that HtrA3 expression was significantly downregulated and the expression of inflammatory cytokines was regulated in response to H/R. HtrA3 overexpression decreased the secretion of inflammatory cytokines, whereas HtrA3 knockdown led to increase levels of inflammatory cytokines. And H/R-induced inflammation in H9C2 cells was inhibited by the regulation of the NF-κB signaling pathway. Our findings demonstrate that HtrA3 alleviates H/R-induced inflammatory responses in H9C2 cardiomyoblasts, possibly by suppressing the pro-inflammatory NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhiming Shen
- Clinical Medical College, Yangzhou University, Yangzhou, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Fei Sun
- Clinical Medical College, Yangzhou University, Yangzhou, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yi Lu
- Clinical Medical College, Yangzhou University, Yangzhou, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Lei Yuan
- Clinical Medical College, Yangzhou University, Yangzhou, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Qian Gong
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Hongcan Shi
- Clinical Medical College, Yangzhou University, Yangzhou, China; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China.
| |
Collapse
|
17
|
Xu Y, Huang Y, Zhang S, Guo L, Wu R, Fang X, Chen X, Xu H, Nie Q. CircDCLRE1C Regulated Lipopolysaccharide-Induced Inflammatory Response and Apoptosis by Regulating miR-214b-3p/STAT3 Pathway in Macrophages. Int J Mol Sci 2022; 23:6822. [PMID: 35743265 PMCID: PMC9224735 DOI: 10.3390/ijms23126822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/12/2022] [Accepted: 06/18/2022] [Indexed: 11/17/2022] Open
Abstract
The immune cell inflammation response is closely related to the occurrence of disease, and much evidence has shown that circular RNAs (circRNAs) play vital roles in the occurrence of disease. However, the biological function and regulatory mechanisms of circRNAs in the immune cell inflammation response remain poorly understood. In this study, we constructed an inflammatory model using lipopolysaccharide (LPS)-stimulated chicken macrophage lines (also known as HD11) to verify the function and mechanism of the novel circDCLRE1C (ID: gga_circ_0001674), which was significantly upregulated in spleen tissues infected by coccidia and the macrophage cells exposed to LPS. The results showed that circDCLRE1C aggravated LPS-induced inflammation and apoptosis in HD11 cells. Systemically, circDCLRE1C acted as a sponge for miR-214b-3p binding sites thereby regulating the expression of STAT3. The overexpression of miR-214b-3p rescued the pro-inflammatory effect of circDCLRE1C in HD11 cells stimulated with LPS, and rescued the high expression of STAT3. In conclusion, our study showed that circDCLRE1C could aggravate LPS-induced inflammation and apoptosis through competitive adsorption of miR-214b-3p, thereby increasing the expression of STAT3.
Collapse
Affiliation(s)
- Yibin Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yulin Huang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siyu Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Lijin Guo
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Ruiquan Wu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiang Fang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaolan Chen
- School of Life Sciences, Chongqing University, Chongqing 401331, China;
| | - Haiping Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qinghua Nie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Y.X.); (Y.H.); (S.Z.); (L.G.); (R.W.); (X.F.)
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources & Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
18
|
Li W, Li D, Chen Y, Abudou H, Wang H, Cai J, Wang Y, Liu Z, Liu Y, Fan H. Classic Signaling Pathways in Alveolar Injury and Repair Involved in Sepsis-Induced ALI/ARDS: New Research Progress and Prospect. DISEASE MARKERS 2022; 2022:6362344. [PMID: 35726235 PMCID: PMC9206211 DOI: 10.1155/2022/6362344] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022]
Abstract
Sepsis is a common critical clinical disease with high mortality that can cause approximately 10 million deaths worldwide each year. Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a common clinical complication of sepsis, which occurs primarily as diffuse alveolar injury, hypoxemia, and respiratory distress. The mortality rate of ALI/ARDS is as high as 30%-40%, which greatly endangers human health. Due to the unclear pathogenesis of ALI/ARDS, its treatment is still a worldwide problem. At present, clinical treatment mainly relies on lung-protective ventilation, prone position ventilation, and fluid management. However, there is a lack of effective and specific treatment measures. In recent years, domestic and foreign scholars have committed to basic research on ALI/ARDS, trying to further clarify its pathogenesis and find new targets and methods for the treatment of ALI/ARDS. In this review, we summarize the signaling pathways related to alveolar injury and repair in sepsis-induced ALI/ARDS and their latest research progress. They include the NF-κB, JAK2/STAT3, mitogen-activated protein kinase (MAPK), mTOR, and Notch signaling pathways. Understanding the molecular mechanisms of these signaling pathways in sepsis-induced ALI/ARDS may provide new targets and ideas for the clinical treatment of this disease.
Collapse
Affiliation(s)
- Wenli Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Duo Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yuansen Chen
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Halidan Abudou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Haiwang Wang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Jinxia Cai
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yiping Wang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Ziquan Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yanqing Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| |
Collapse
|
19
|
Dou T, Wang J, Liu Y, Jia J, Zhou L, Liu G, Li X, Han M, Lin J, Huang F, Chen X. A Combined Transcriptomic and Proteomic Approach to Reveal the Effect of Mogroside V on OVA-Induced Pulmonary Inflammation in Mice. Front Immunol 2022; 13:800143. [PMID: 35371026 PMCID: PMC8972588 DOI: 10.3389/fimmu.2022.800143] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
Mogroside V is a bioactive ingredient extracted from the natural food Siraitia grosvenorii which possesses functions that stimulate lung humidification and cough relief activities, but its underlying mechanisms were rarely studied. To estimate its potential protective effect on ovalbumin (OVA)-induced pulmonary inflammation and understand its system-wide mechanism, integrated omics was applied in this study. Mogroside V effectively reduced the levels of IgE, TNF-α, and IL-5 in OVA-induced mice. The results of RNA-seq and data-independent acquisition proteomics approach revealed that 944 genes and 341 proteins were differentially expressed in the normal control group (NC) and ovalbumin-induced control group (OC) and 449 genes and 259 proteins were differentially expressed between the OC and the group treated with 50 mg/kg mogroside V (MV). After a combined analysis of the transcriptome and the proteome, 93 major pathways were screened, and we discovered that mogroside V exerts an anti-inflammation effect in the lung via NF-κB and JAK-STAT, both of which are among the signaling pathways mentioned above. In addition, we found that the key regulatory molecules (Igha, Ighg1, NF-κB, Jak1, and Stat1) in the two pathways were activated in inflammation and inhibited by mogroside V. Thus, mogroside V may be the main bioactivity component in S. grosvenorii that exerts lung humidification and cough relief effects.
Collapse
Affiliation(s)
- Tong Dou
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Juan Wang
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, The Affiliated Hospital of Guilin Medical University, Guilin, China
- Faculty of Basic Medicine, Guilin Medical University, Guilin, China
| | - Yisa Liu
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Jiangang Jia
- Department of Pharmacy, Guilin Medical University, Guilin, China
| | - Luwei Zhou
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Guoxiang Liu
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Xiaojuan Li
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Mengjie Han
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Jiaxun Lin
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Fengxiang Huang
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Xu Chen
- Department of Pharmacy, Guilin Medical University, Guilin, China
- Key Laboratory of Pharmacognosy, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- *Correspondence: Xu Chen,
| |
Collapse
|
20
|
Gao Y, Li S, Dong R, Li X. Long noncoding RNA MIR3142HG accelerates lipopolysaccharide-induced acute lung injury via miR-95-5p/JAK2 axis. Hum Cell 2022; 35:856-870. [DOI: 10.1007/s13577-022-00687-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/22/2022] [Indexed: 12/01/2022]
|
21
|
Cao J, Liu M, Feng S, Li Y, Zheng K. Glaucocalyxin A alleviates lipopolysaccharide‑induced inflammation and apoptosis in pulmonary microvascular endothelial cells and permeability injury by inhibiting STAT3 signaling. Exp Ther Med 2022; 23:313. [PMID: 35369532 PMCID: PMC8943557 DOI: 10.3892/etm.2022.11242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/29/2021] [Indexed: 11/05/2022] Open
Abstract
Glaucocalyxin A (GLA), an ent-kauranoid diterpene derived from Rabdosia japonica var. glaucocalyx, possesses antibacterial, anti-oxidative and anti-neuroinflammatory properties. The present study aimed to investigate the potential mechanisms underlying GLA in the pathogenesis of pneumonia. Human pulmonary microvascular endothelial cells (hPMVECs) treated with lipopolysaccharide (LPS) were treated with GLA, followed by the detection of cell viability, inflammation, apoptosis and cell permeability. Furthermore, the protein expression levels of apoptosis- and permeability-associated proteins were determined using western blot analysis. Following treatment with a signal transducer and activator of transcription 3 (STAT3) activator, the protein expression levels of STAT3 and endoplasmic reticulum stress-associated proteins were determined, to confirm whether STAT3 signaling was mediated by GLA. Lastly, the mRNA expression level of inflammatory cytokines, apoptosis and permeability injury were also determined following treatment with a STAT3 activator. The results revealed that GLA ameliorated inflammation, apoptosis and permeability injury in LPS-induced hPMVECs. Following treatment with a STAT3 activator, the therapeutic effects of GLA on LPS-induced hPMVECs were abrogated. In conclusion, GLA alleviated LPS-induced inflammation, apoptosis and permeability injury in hPMVECs by inhibiting STAT3 signaling, which highlighted the potential therapeutic value of GLA in the treatment of pneumonia.
Collapse
Affiliation(s)
- Jianwei Cao
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Meiling Liu
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Shufang Feng
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Yingying Li
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| | - Kaijun Zheng
- Pediatrics Department, Zhongshan People's Hospital, Zhongshan, Guangdong 528403, P.R. China
| |
Collapse
|
22
|
Al‐Ani B, ShamsEldeen AM, Kamar SS, Haidara MA, Al‐Hashem F, Alshahrani MY, Al‐Hakami AM, Kader DHA, Maarouf A. Lipopolysaccharide induces acute lung injury and alveolar hemorrhage in association with the cytokine storm, coagulopathy and AT1R/JAK/STAT augmentation in a rat model that mimics moderate and severe Covid‐19 pathology. Clin Exp Pharmacol Physiol 2022; 49:483-491. [PMID: 35066912 DOI: 10.1111/1440-1681.13620] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Bahjat Al‐Ani
- Department of Physiology College of Medicine King Khalid University Abha 61421 Saudi Arabia
| | - Asmaa M. ShamsEldeen
- Department of Physiology Kasr Al‐Aini Faculty of Medicine Cairo University Cairo Egypt
| | - Samaa S. Kamar
- Department of Medical Histology Kasr Al‐Aini Faculty of Medicine Cairo University Cairo Egypt
| | - Mohamed A. Haidara
- Department of Physiology Kasr Al‐Aini Faculty of Medicine Cairo University Cairo Egypt
| | - Fahaid Al‐Hashem
- Department of Physiology College of Medicine King Khalid University Abha 61421 Saudi Arabia
| | - Mohammad Y. Alshahrani
- Research Center for Advanced Materials Science (RCAMS) King Khalid University Abha 61413 Saudi Arabia
- Department of Clinical Laboratory Sciences College of Applied Medical Sciences King Khalid University Abha 61413 Saudi Arabia
| | - Ahmed M. Al‐Hakami
- Department of Microbiology and Clinical Parasitology College of Medicine King Khalid University Abha 61421 Saudi Arabia
| | - Dina H. Abdel Kader
- Department of Medical Histology Kasr Al‐Aini Faculty of Medicine Cairo University Cairo Egypt
| | - Amro Maarouf
- Department of Clinical Biochemistry University Hospitals Birmingham NHS Foundation Trust Birmingham UK
| |
Collapse
|
23
|
Kiszałkiewicz JM, Majewski S, Piotrowski WJ, Górski P, Pastuszak-Lewandoska D, Migdalska-Sęk M, Brzeziańska-Lasota E. Evaluation of selected IL6/STAT3 pathway molecules and miRNA expression in chronic obstructive pulmonary disease. Sci Rep 2021; 11:22756. [PMID: 34815425 PMCID: PMC8610981 DOI: 10.1038/s41598-021-01950-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
COPD has been regarded as a global epidemic due to an increase in pollution and tobacco exposure. Therefore, the study of molecular mechanism as the basis for modern therapy is important. The aim of the study was the assessment of gene expression levels, IL-6, IL-6ST, PIAS3, STAT3, and miRNAs, miRNA-1, miRNA-106b, miRNA-155, in patients with COPD. Induced sputum as well as PBMC were collected from 40 patients clinically verified according to the GOLD 2021 (A-D) classification and from the control group (n = 20). The levels of gene and miRNA expression were analysed by qPCR. In induced sputum IL6 was significantly down-regulated in COPD group compared with control (p = 0.0008), while IL6ST were up-regulated (p = 0.05). The results were also statistically significant for STAT3 (p = 0.04) and miRNA-155 (p = 0.03) with higher expression in the current smokers compared to ex-smokers. Higher expression levels for IL6ST (p = 0.03) in COPD patients with the exacerbation history compared to COPD patients without the exacerbation history were noted. Compared induced sputum and PB lymphocytes we observed higher expression of IL6 (p = 0.0003), STAT3 (p = 0.000001) miRNA-106b (p = 0.000069 and miRNA-155 (p = 0.000016) in induced sputum with lower expression of PIAS3 (p = 0.006), IL6ST (p = 0.002) and miRNA-1 (p = 0.001). Differences in gene expression levels of the IL-6/IL6ST/STAT3 pathway and miRNA depending on the smoking status and classification of patients according to GOLD suggest the importance of these genes in the pathogenesis of COPD and may indicate their potential utility in monitoring the course of the disease.
Collapse
Affiliation(s)
- J M Kiszałkiewicz
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, St. Pomorska 251, 92-213, Lodz, Poland.
| | - S Majewski
- Department of Pneumology, Medical University of Lodz, St. Kopcińskiego 22, 90-153, Lodz, Poland
| | - W J Piotrowski
- Department of Pneumology, Medical University of Lodz, St. Kopcińskiego 22, 90-153, Lodz, Poland
| | - P Górski
- Department of Pneumology, Medical University of Lodz, St. Kopcińskiego 22, 90-153, Lodz, Poland
| | - D Pastuszak-Lewandoska
- Department of Microbiology and Laboratory Medical Immunology, Chair of Biology and Medical Microbiology, Medical University of Lodz, St. Pomorska 251, 92-213, Lodz, Poland
| | - M Migdalska-Sęk
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, St. Pomorska 251, 92-213, Lodz, Poland
| | - E Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, St. Pomorska 251, 92-213, Lodz, Poland
| |
Collapse
|
24
|
Zhang X, Zheng C, Gao Z, Wang L, Chen C, Zheng Y, Meng Y. PKM2 promotes angiotensin-II-induced cardiac remodelling by activating TGF-β/Smad2/3 and Jak2/Stat3 pathways through oxidative stress. J Cell Mol Med 2021; 25:10711-10723. [PMID: 34687136 PMCID: PMC8581335 DOI: 10.1111/jcmm.17007] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/24/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
Hypertensive cardiac remodelling is a common cause of heart failure. However, the molecular mechanisms regulating cardiac remodelling remain unclear. Pyruvate kinase isozyme type M2 (PKM2) is a key regulator of the processes of glycolysis and oxidative phosphorylation, but the roles in cardiac remodelling remain unknown. In the present study, we found that PKM2 was enhanced in angiotensin II (Ang II)-treated cardiac fibroblasts and hypertensive mouse hearts. Suppression of PKM2 by shikonin alleviated cardiomyocyte hypertrophy and fibrosis in Ang-II-induced cardiac remodelling in vivo. Furthermore, inhibition of PKM2 markedly attenuated the function of cardiac fibroblasts including proliferation, migration and collagen synthesis in vitro. Mechanistically, suppression of PKM2 inhibited cardiac remodelling by suppressing TGF-β/Smad2/3, Jak2/Stat3 signalling pathways and oxidative stress. Together, this study suggests that PKM2 is an aggravator in Ang-II-mediated cardiac remodelling. The negative modulation of PKM2 may provide a promising therapeutic approach for hypertensive cardiac remodelling.
Collapse
Affiliation(s)
- Xiyu Zhang
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Beijing Lab for Cardiovascular Precision Medicine, Department of Pathology, Capital Medical University, Beijing, China
| | - Cuiting Zheng
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Beijing Lab for Cardiovascular Precision Medicine, Department of Pathology, Capital Medical University, Beijing, China
| | - Zhenqiang Gao
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Beijing Lab for Cardiovascular Precision Medicine, Department of Pathology, Capital Medical University, Beijing, China
| | - Lingling Wang
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chen Chen
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Zheng
- Department of Pharmacology, Capital Medical University, Beijing, China
| | - Yan Meng
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Beijing Lab for Cardiovascular Precision Medicine, Department of Pathology, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
The Effect of Resveratrol on the Cardiovascular System from Molecular Mechanisms to Clinical Results. Int J Mol Sci 2021; 22:ijms221810152. [PMID: 34576315 PMCID: PMC8466271 DOI: 10.3390/ijms221810152] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases are the leading causes of death worldwide. The cardioprotective effects of natural polyphenols such as resveratrol (3,5,4-trihydroxystilbene) have been extensively investigated throughout recent decades. Many studies of RES have focused on its favorable effects on pathological conditions related to cardiovascular diseases and their risk factors. The aim of this review was to summarize the wide beneficial effects of resveratrol on the cardiovascular system, including signal transduction pathways of cell longevity, energy metabolism of cardiomyocytes or cardiac remodeling, and its anti-inflammatory and antioxidant properties. In addition, this paper discusses the significant preclinical and human clinical trials of recent years with resveratrol on cardiovascular system. Finally, we present a short overview of antiviral and anti-inflammatory properties and possible future perspectives on RES against COVID-19 in cardiovascular diseases.
Collapse
|
26
|
Daphnetin ameliorates acute lung injury in mice with severe acute pancreatitis by inhibiting the JAK2-STAT3 pathway. Sci Rep 2021; 11:11491. [PMID: 34075160 PMCID: PMC8169853 DOI: 10.1038/s41598-021-91008-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Severe acute pancreatitis (SAP) is often associated with pulmonary inflammation leading to acute lung injury. Daphnetin, a natural coumarin derivative, has been reported to exert anti-inflammatory effects. Here, we explored the effect and possible mechanism of daphnetin in a mouse model of SAP-associated lung injury induced by an intraperitoneal injection of L-arginine. The severity of pancreatic and lung injury is determined by histology and its score. Immunostaining of inflammatory and apoptotic cells was used to demonstrate lung tissue inflammation and apoptosis; ELISA analysis of serum and tissue cytokine levels; and western blotting and immunohistochemical staining for the activated Janus kinase 2 (JAK2)-signal transducer and activator of transcription protein 3 (STAT3) signalling pathway in lung tissues. Daphnetin pretreatment significantly reduced SAP-induced pancreatic and lung tissue damage, reduced interleukin-6 and tumour necrosis factor-α concentrations in both serum and lung tissues, reduced serum amylase and myeloperoxidase activities, and reduced macrophage (CD11b) and neutrophil (Ly6G) infiltration and cell apoptosis in the lung tissue. Moreover, SAP-induced phosphorylation of JAK2 and STAT3 in the lung tissue was also significantly diminished by the daphnetin pretreatment. These results indicated that daphnetin reduces SAP-associated lung tissue damage, likely by inhibiting the activation of JAK2-STAT3 signalling.
Collapse
|
27
|
Esculetin protects against early sepsis via attenuating inflammation by inhibiting NF-κB and STAT1/STAT3 signaling. Chin J Nat Med 2021; 19:432-441. [PMID: 34092294 DOI: 10.1016/s1875-5364(21)60042-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Indexed: 01/14/2023]
Abstract
Esculetin, a natural derivative from the traditional and widely-used Chinese medicinal herb Cortex Fraxini, has a variety of pharmacological effects, especially in anti-inflammation. However, it is not clear whether esculetin has a therapeutic effect on sepsis. This study aimed to investigate the anti-inflammatory and protective effects of esculetin on early sepsis. The results showed that the lung injury was significantly relieved with the treatment of esculetin, accompanied with the restrained production of inflammatory factors including IL-1β, IL-6, TNF-α, CCL2 and iNOS during the early phase of E.coli-induced sepsis. Of note, activation of NF-κB and STAT1/STAT3 signals, the main upstream signals of many inflammatory factors, were attenuated by esculetin in both lung tissues from septic mice and LPS-stimulated macrophage. These findings suggested that the protection of esculetin against early sepsis should be related to its anti-inflammatory effect, which was at least partly due to its inhibition on NF-κB and STAT1/STAT3 signaling pathway in macrophage. Thus, esculetin could serve as a potential therapeutic agent by rebalancing innate immune response in macrophage for the treatment of early sepsis.
Collapse
|
28
|
Da Cheng Qi Decoction Alleviates Cerulein-Stimulated AR42J Pancreatic Acinar Cell Injury via the JAK2/STAT3 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6657036. [PMID: 33927777 PMCID: PMC8053057 DOI: 10.1155/2021/6657036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/08/2021] [Accepted: 03/27/2021] [Indexed: 12/22/2022]
Abstract
Background Acute pancreatitis (AP) is a common acute abdomen inflammation, characterized by the dysregulation of digestive enzyme production and secretion. Many studies have shown that Da Cheng Qi Decoction (DCQD) is a secure, effective prescription on AP. In this study, cerulein-stimulated AR42J cells damage model was established to further explore the feasibility and underlying mechanism of DCQD as a potential inhibitor of JAK2/STAT3 pathway for the treatment of AP. Methods Cell viability of DCQD was measured using a cell counting Kit-8 assay. Pancreatic biochemical markers such as amylase, lipase, and C-reactive protein production were measured by assay kits, respectively. Cytokines (TNF-α, IL-6, IL-10, and IL-1β) were assayed by ELISA. Protein location and protein expression were detected by immunofluorescence staining and Western blotting, respectively. Gene expression was assessed by real-time PCR. For mechanistic analysis of the effect of DCQD on JAK2/STAT3 signaling pathway, selective JAK2 inhibitor (Fedratinib) and STAT3 inhibitor (Stattic) as well as STAT3 activator (Garcinone D) were used. Results DCQD protected cells by regulating cerulein-induced inflammation and reducing the secretion of pancreatic biochemical markers. Moreover, DCQD could not only inhibit the nuclear translocation of p-STAT3, but also decrease the mRNA expression of JAK2 and STAT3 as well as the ratio of p-JAK2/JAK2 and p-STAT3/STAT3 in protein level. Additionally, DCQD could regulate the mRNA and protein expression of JAK2/STAT3 downstream effectors, Bax and Bcl-XL. The activated effect of cerulein on JAK2/STAT3 pathway was also reversed by JAK2 inhibitor Fedratinib or STAT3 inhibitor Stattic. And the overexpression of JAK2/STAT3 pathway, via STAT3 activator Garcinone D, did exert damage on cells, which bore a resemblance to cerulein. Conclusion The activation of JAK2/STAT3 pathway may play a key role in the pathogenesis of cerulein-stimulated AR42J pancreatic acinar cell injury. DCQD could improve inflammatory cytokines and cell injury, which might be mediated by suppressing the activation of JAK2/STAT3 signaling pathway.
Collapse
|
29
|
Activation of the IL-1β/KLF2/HSPH1 pathway promotes STAT3 phosphorylation in alveolar macrophages during LPS-induced acute lung injury. Biosci Rep 2021; 40:222192. [PMID: 32091104 PMCID: PMC7056450 DOI: 10.1042/bsr20193572] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) is a lethal disease with diffuse lung inflammation, in which JAK/STAT3 signaling has been well recognized for its role in initiating and amplifying inflammatory processes. However, the mechanism for the enhancement and maintenance of signal transducer and activator of transcription 3 (STAT3) activation has not yet been clearly demonstrated in ALI. In the present work, we established a lipopolysaccharide (LPS)-induced ALI rat model through intratracheal instillation and isolated the alveolar macrophages (AMs) from the rats in the model. We demonstrated that the expression of Kruppel-like factor 2 (KLF2) significantly decreased in the AMs from LPS-induced ALI rats (LPS-AMs) as compared with the AMs from control rats (NC-AMs). Overexpressing KLF2 in LPS-AMs inhibited the phosphorylation of STAT3 and reduced the levels of STAT3 target genes, including matrix metalloproteinase (MMP)-2/9 (MMP-2/9). Further investigation indicated that KLF2 trans-inhibited heat shock protein H1 (HSPH1), which interacted with STAT3 and enhanced its phosphorylation. As a crucial inflammatory mediator in ALI, interleukin-1β (IL-1β) induced the down-regulation of KLF2 in LPS-AMs, as interrupting IL-1β signaling in LPS-AMs by antibody neutralization or IL1R1 knockdown rescued the expression of KLF2. Consistently, stimulating NC-AMs with IL-1β decreased KLF2 and increased HSPH1, while overexpression of KLF2 suppressed IL-1β-induced HSPH1. Additionally, in vivo studies showed that treatment with an IL-1β antibody or HSPH1 inhibitor alleviated lung injury in ALI rats, as well as decreased the levels of p-STAT3 and MMP-2/9. In conclusion, activation of the IL-1β/KLF2/HSPH1 pathway facilitated STAT3 phosphorylation in AMs, which exacerbated pulmonary inflammation in ALI.
Collapse
|
30
|
Yun Y, Chen J, Wang X, Li Y, Hu Z, Yang P, Qin L. Tofacitinib Ameliorates Lipopolysaccharide-Induced Acute Kidney Injury by Blocking the JAK-STAT1/STAT3 Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8877056. [PMID: 33511217 PMCID: PMC7822662 DOI: 10.1155/2021/8877056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/05/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Septic acute kidney injury (AKI) is the most common AKI syndrome in the intensive care unit (ICU), and it accounts for approximately half of AKI cases. Tofacitinib (TOFA) is a pan-Janus kinase (JAK) inhibitor that exhibits potent anti-inflammatory activity in rheumatoid arthritis. However, no study has examined the functional role of TOFA in septic AKI. In the present study, we investigated the protective effects of TOFA on septic AKI and the underlying mechanisms. A lipopolysaccharide- (LPS-) induced AKI model was established in C57BL/6 mice via an intraperitoneal injection of LPS (10 mg/kg). One hour after LPS challenge, the mice were orally administered TOFA (5, 10, or 15 mg/kg) every 6 h until sacrifice at 24 h. We found that TOFA significantly ameliorated LPS-induced renal histopathological changes and dysfunction. TOFA also suppressed the expression levels of proinflammatory cytokines (TNF-α, IL-1β, IL-6, and IFN-γ) and the parameters of oxidative stress (MDA, GSH, SOD, and CAT) in kidney tissues. These results may be associated with the inhibitory effect of TOFA on the JAK-STAT1/STAT3 pathway, which was significantly activated by LPS challenge. TOFA treatment also inhibited LPS-induced activation of the TLR4/NF-κB pathway. In conclusion, we revealed that TOFA had a protective effect on LPS-induced AKI, and it may be a promising therapeutic agent for septic AKI.
Collapse
Affiliation(s)
- Yang Yun
- Department of Physiology, China Medical University, Liaoning 110122, China
| | - Jingyu Chen
- Department of Physiology, China Medical University, Liaoning 110122, China
| | - Xuejiao Wang
- Department of Physiology, China Medical University, Liaoning 110122, China
| | - Yingzhuo Li
- Department of Physiology, China Medical University, Liaoning 110122, China
| | - Zhifan Hu
- Department of Physiology, China Medical University, Liaoning 110122, China
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Liaoning 110001, China
| | - Ling Qin
- Department of Physiology, China Medical University, Liaoning 110122, China
| |
Collapse
|
31
|
Wan X, Tian X, Du J, Lu Y, Xiao Y. Long non-coding RNA H19 deficiency ameliorates bleomycin-induced pulmonary inflammation and fibrosis. Respir Res 2020; 21:290. [PMID: 33138822 PMCID: PMC7607673 DOI: 10.1186/s12931-020-01534-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022] Open
Abstract
Background The poor understanding of pathogenesis in idiopathic pulmonary fibrosis (IPF) impaired development of effective therapeutic strategies. The aim of the current study is to investigate the roles of long non-coding RNA H19 (lncRNA H19) in the pulmonary inflammation and fibrosis of IPF. Methods Bleomycin was used to induce pulmonary inflammation and fibrosis in mice. The mRNAs and proteins expression in lung tissues was determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. H19 knockout (H19−/−) mice were generated by CRISPR/Cas9. Results The expression of H19 mRNA was up-regulated in fibrotic lungs patients with IPF as well as in lungs tissues that obtained from bleomycin-treated mice. H19−/− mice suppressed bleomycin-mediated pulmonary inflammation and inhibited the Il6/Stat3 signaling. H19 deficiency ameliorated bleomycin-induced pulmonary fibrosis and repressed the activation of TGF-β/Smad and S1pr2/Sphk2 in the lungs of bleomycin-treated mice. Conclusions Our data suggests that H19 is a profibrotic lncRNA and a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Xiaoyu Wan
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xinbei Tian
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Jun Du
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Ying Lu
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Yongtao Xiao
- Shanghai Institute for Pediatric Research, Shanghai, China. .,Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kong Jiang Road, Shanghai, China.
| |
Collapse
|
32
|
Xue H, Li M. Protective effect of pterostilbene on sepsis-induced acute lung injury in a rat model via the JAK2/STAT3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1452. [PMID: 33313197 PMCID: PMC7723647 DOI: 10.21037/atm-20-5814] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Bacterial infection is one of the most common causes of sepsis, with acute lung injury (ALI) being a related complication. Pterostilbene (PTS) is extracted from blueberries, peanuts, and grapes, and has numerous pharmacologic activities. The aim of the present study was to explore the underlying role of PTS protects against sepsis-mediated ALI. Methods We established a sepsis model induced by cecal ligation and puncture (CLP) in rats. The rats were randomly divided into five groups (n=5 each): sham group, CLP group, Dexmedetomidine group (Dex, 50 µg/kg) and PTS groups (25 and 50 mg/kg). Twenty-hours hours after CLP, PTS was intraperitoneally injected for 14 continuous days. The rats were killed, and blood and lung tissue were collected for pathological analysis and mRNA and protein detection. Results Our findings showed that PTS reduced the wet/dry ratio and ameliorated sepsis-induced pulmonary fibrosis (PF), which was associated with improvement of pathological damage in lung tissues. We also observed the inhibitory effect of PTS on apoptosis and release of inflammatory cytokines (i.e., tumor necrosis factor-α, interleukin-6, and monocyte chemotactic protein 1). In addition, PTS markedly suppressed the phosphorylation levels of Janus kinase-2 (JAK2) and signal transducer and activator of transcription 3 (STAT3). Conclusions Our results indicated that PTS inhibited the PF, apoptosis, and inflammatory response via the JAK2/STAT3 pathway in a sepsis-induced ALI rat model, providing a candidate for drug therapy of sepsis-induced ALI.
Collapse
Affiliation(s)
- Hua Xue
- Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
33
|
A Perspective on Erythropoietin as a Potential Adjuvant Therapy for Acute Lung Injury/Acute Respiratory Distress Syndrome in Patients with COVID-19. Arch Med Res 2020; 51:631-635. [PMID: 32863034 PMCID: PMC7418647 DOI: 10.1016/j.arcmed.2020.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
Abstract
The novel coronavirus 2019-nCoV (SARS-CoV-2) infection that emerged in China in December 2019 has rapidly spread to become a global pandemic. This article summarizes the potential benefits of erythropoietin (EPO) in alleviating SARS-CoV-2 pathogenesis which is now called COVID-19. As with other coronavirus infection, the lethality of COVID-19 is associated with respiratory dysfunction due to overexpression of proinflammatory cytokines induced by the host immune responses. The resulting cytokine storm leads to the development of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Erythropoietin, well known for its role in the regulation of erythropoiesis, may have protective effects against ALI/ARDS induced by viral and other pathogens. EPO exerts antiapoptotic and cytoprotective properties under various pathological conditions. With a high safety profile, EPO promotes the production of endothelial progenitor cells and reduce inflammatory processes through inhibition of the nuclear factor-κB (NF-κB) and JAK-STAT3 signaling pathways. Thus, it may be considered as a safe drug candidate for COVID-19 patients if given at the early stage of the disease. The potential effects of erythropoietin on different aspects of ALI/ARDS associated with SARS-CoV-2 infection are reviewed.
Collapse
|
34
|
Paris AJ, Hayer KE, Oved JH, Avgousti DC, Toulmin SA, Zepp JA, Zacharias WJ, Katzen JB, Basil MC, Kremp MM, Slamowitz AR, Jayachandran S, Sivakumar A, Dai N, Wang P, Frank DB, Eisenlohr LC, Cantu E, Beers MF, Weitzman MD, Morrisey EE, Worthen GS. STAT3-BDNF-TrkB signalling promotes alveolar epithelial regeneration after lung injury. Nat Cell Biol 2020; 22:1197-1210. [PMID: 32989251 PMCID: PMC8167437 DOI: 10.1038/s41556-020-0569-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/03/2020] [Indexed: 01/13/2023]
Abstract
Alveolar epithelial regeneration is essential for recovery from devastating lung diseases. This process occurs when type II alveolar pneumocytes (AT2 cells) proliferate and transdifferentiate into type I alveolar pneumocytes (AT1 cells). We used genome-wide analysis of chromatin accessibility and gene expression following acute lung injury to elucidate repair mechanisms. AT2 chromatin accessibility changed substantially following injury to reveal STAT3 binding motifs adjacent to genes that regulate essential regenerative pathways. Single-cell transcriptome analysis identified brain-derived neurotrophic factor (Bdnf) as a STAT3 target gene with newly accessible chromatin in a unique population of regenerating AT2 cells. Furthermore, the BDNF receptor tropomyosin receptor kinase B (TrkB) was enriched on mesenchymal alveolar niche cells (MANCs). Loss or blockade of AT2-specific Stat3, Bdnf or mesenchyme-specific TrkB compromised repair and reduced Fgf7 expression by niche cells. A TrkB agonist improved outcomes in vivo following lung injury. These data highlight the biological and therapeutic importance of the STAT3-BDNF-TrkB axis in orchestrating alveolar epithelial regeneration.
Collapse
Affiliation(s)
- Andrew J Paris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharina E Hayer
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joseph H Oved
- Division of Hematology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daphne C Avgousti
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sushila A Toulmin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jarod A Zepp
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William J Zacharias
- Division of Pulmonary Biology, Perinatal Institute, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeremy B Katzen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria C Basil
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madison M Kremp
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sowmya Jayachandran
- Division of Cardiology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aravind Sivakumar
- Division of Cardiology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ning Dai
- Division of Neonatology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ping Wang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Cardiology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laurence C Eisenlohr
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Edward Cantu
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael F Beers
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew D Weitzman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Edward E Morrisey
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for Regenerative Medicine, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - G Scott Worthen
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Neonatology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Xiu MX, Liu ZT, Tang J. Screening and identification of key regulatory connections and immune cell infiltration characteristics for lung transplant rejection using mucosal biopsies. Int Immunopharmacol 2020; 87:106827. [PMID: 32791489 PMCID: PMC7417178 DOI: 10.1016/j.intimp.2020.106827] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/03/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
This study aimed to explore key regulatory connections underlying lung transplant rejection. The differentially expressed genes (DEGs) between rejection and stable lung transplantation (LTx) samples were screened using R package limma, followed by functional enrichment analysis and protein-protein interaction network construction. Subsequently, a global triple network, including miRNAs, mRNAs, and transcription factors (TFs), was constructed. Furthermore, immune cell infiltration characteristics were analyzed to investigate the molecular immunology of lung transplant rejection. Finally, potential drug-target interactions were generated. In brief, 739 DEGs were found between rejection and stable LTx samples. PTPRC, IL-6, ITGAM, CD86, TLR8, TYROBP, CXCL10, ITGB2, and CCR5 were defined as hub genes. Eight TFs, including STAT1, SPIB, NFKB1, SPI1, STAT5A, RUNX1, VENTX, and BATF, and five miRNAs, including miR-335-5p, miR-26b-5p, miR-124-3p, miR-1-3p, and miR-155-5p, were involved in regulating hub genes. The immune cell infiltration analysis revealed higher proportions of activated memory CD4 T cells, follicular helper T cells, γδ T cells, monocytes, M1 and M2 macrophages, and eosinophils in rejection samples, besides lower proportions of resting memory CD4 T cells, regulatory T cells, activated NK cells, M0 macrophages, and resting mast cells. This study provided a comprehensive perspective of the molecular co-regulatory network underlying lung transplant rejection.
Collapse
Affiliation(s)
- Meng-Xi Xiu
- Medical School of Nanchang University, Nanchang, PR China
| | - Zu-Ting Liu
- Medical School of Nanchang University, Nanchang, PR China
| | - Jian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
36
|
Beneficial effect of STAT3 decoy oligodeoxynucleotide transfection on organ injury and mortality in mice with cecal ligation and puncture-induced sepsis. Sci Rep 2020; 10:15316. [PMID: 32943679 PMCID: PMC7498613 DOI: 10.1038/s41598-020-72136-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 08/24/2020] [Indexed: 12/25/2022] Open
Abstract
Sepsis is a major clinical challenge with unacceptably high mortality. The signal transducers and activators of transcription (STAT) family of transcription factors is known to activate critical mediators of cytokine responses, and, among this family, STAT3 is implicated to be a key transcription factor in both immunity and inflammatory pathways. We investigated whether in vivo introduction of synthetic double-stranded STAT3 decoy oligodeoxynucleotides (ODNs) can provide benefits for reducing organ injury and mortality in mice with cecal ligation and puncture (CLP)-induced polymicrobial sepsis. We found that STAT3 was rapidly activated in major end-organ tissues following CLP, which was accompanied by activation of the upstream kinase JAK2. Transfection of STAT3 decoy ODNs downregulated pro-inflammatory cytokine/chemokine overproduction in CLP mice. Moreover, STAT3 decoy ODN transfection significantly reduced the increases in tissue mRNAs and proteins of high mobility group box 1 (HMGB1) and strongly suppressed the excessive elevation in serum HMGB1 levels in CLP mice. Finally, STAT3 decoy ODN administration minimized the development of sepsis-driven major end-organ injury and led to a significant survival advantage in mice after CLP. Our results suggest a critical role of STAT3 in the sepsis pathophysiology and the potential usefulness of STAT3 decoy ODNs for sepsis gene therapy.
Collapse
|
37
|
Battagello D, Dragunas G, Klein M, Ayub AL, Velloso F, Correa R. Unpuzzling COVID-19: tissue-related signaling pathways associated with SARS-CoV-2 infection and transmission. Clin Sci (Lond) 2020; 134:2137-2160. [PMID: 32820801 PMCID: PMC7443512 DOI: 10.1042/cs20200904] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
The highly infective coronavirus disease 19 (COVID-19) is caused by a novel strain of coronaviruses - the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) - discovered in December 2019 in the city of Wuhan (Hubei Province, China). Remarkably, COVID-19 has rapidly spread across all continents and turned into a public health emergency, which was ultimately declared as a pandemic by the World Health Organization (WHO) in early 2020. SARS-CoV-2 presents similar aspects to other members of the coronavirus family, mainly regarding its genome, protein structure and intracellular mechanisms, that may translate into mild (or even asymptomatic) to severe infectious conditions. Although the mechanistic features underlying the COVID-19 progression have not been fully clarified, current evidence have suggested that SARS-CoV-2 may primarily behave as other β-coronavirus members. To better understand the development and transmission of COVID-19, unveiling the signaling pathways that may be impacted by SARS-CoV-2 infection, at the molecular and cellular levels, is of crucial importance. In this review, we present the main aspects related to the origin, classification, etiology and clinical impact of SARS-CoV-2. Specifically, here we describe the potential mechanisms of cellular interaction and signaling pathways, elicited by functional receptors, in major targeted tissues/organs from the respiratory, gastrointestinal (GI), cardiovascular, renal, and nervous systems. Furthermore, the potential involvement of these signaling pathways in evoking the onset and progression of COVID-19 symptoms in these organ systems are presently discussed. A brief description of future perspectives related to potential COVID-19 treatments is also highlighted.
Collapse
Affiliation(s)
- Daniella S. Battagello
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Guilherme Dragunas
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Marianne O. Klein
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Ana L.P. Ayub
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Fernando J. Velloso
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-NJMS, Newark, NJ, U.S.A
| | - Ricardo G. Correa
- NCI-Designated Cancer Center, Sanford Burnham Prebys (SBP) Medical Discovery Institute, La Jolla, CA, U.S.A
| |
Collapse
|
38
|
Thomas G, Frederick E, Hausburg M, Goldberg L, Hoke M, Roshon M, Mains C, Bar-Or D. The novel immunomodulatory biologic LMWF5A for pharmacological attenuation of the "cytokine storm" in COVID-19 patients: a hypothesis. Patient Saf Surg 2020; 14:21. [PMID: 32431755 PMCID: PMC7220573 DOI: 10.1186/s13037-020-00248-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A common complication of viral pulmonary infections, such as in the ongoing COVID-19 pandemic, is a phenomenon described as a "cytokine storm". While poorly defined, this hyperinflammatory response results in diffuse alveolar damage. The low molecular weight fraction of commercial human serum albumin (LMWF5A), a novel biologic in development for osteoarthritis, demonstrates beneficial in vitro immunomodulatory effects complimentary to addressing inflammation, thus, we hypothesize that LMWF5A could improve the clinical outcomes of COVID-19 by attenuating hyperinflammation and the potential development of a cytokine storm. PRESENTATION OF THE HYPOTHESIS A variety of human in vitro immune models indicate that LMWF5A reduces the production of pro-inflammatory cytokines implicated in cytokine storm associated with COVID-19. Furthermore, evidence suggests LMWF5A also promotes the production of mediators required for resolving inflammation and enhances the barrier function of endothelial cultures. TESTING THE HYPOTHESIS A randomized controlled trial, to evaluate the safety and efficacy of nebulized LMWF5A in adults with Acute Respiratory Distress Syndrome (ARDS) secondary to COVID-19 infection, was developed and is currently under review by the Food and Drug Administration. IMPLICATIONS OF HYPOTHESIS If successful, this therapy may attenuate the cytokine storm observed in these patients and potentially reduce mortality, increase ventilation free days, improve oxygenation parameters and consequently lessen the burden on patients and the intensive care unit. CONCLUSIONS In conclusion, in vitro findings suggest that the immunomodulatory effects of LMWF5A make it a viable candidate for treating cytokine storm and restoring homeostasis to the immune response in COVID-19.
Collapse
Affiliation(s)
- Gregory Thomas
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Elizabeth Frederick
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Melissa Hausburg
- Trauma Research Department, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113 USA
- Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228 USA
- Trauma Research Department, Medical City Plano, 3901 W 15th St, Plano, TX 75075 USA
- Trauma Research Department, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907 USA
- Trauma Research Department, Research Medical Center, 2316 E Meyer Blvd, Kansas City, MO 64132 USA
- Trauma Research Department, Wesley Medical Center, 550 N Hillside St, Wichita, KS 67214 USA
| | - Laura Goldberg
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Marshall Hoke
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Michael Roshon
- Emergency Department, Penrose Hospital, Colorado Springs, Colorado USA
| | | | - David Bar-Or
- Trauma Research Department, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113 USA
- Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228 USA
- Trauma Research Department, Medical City Plano, 3901 W 15th St, Plano, TX 75075 USA
- Trauma Research Department, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907 USA
- Trauma Research Department, Research Medical Center, 2316 E Meyer Blvd, Kansas City, MO 64132 USA
- Trauma Research Department, Wesley Medical Center, 550 N Hillside St, Wichita, KS 67214 USA
- Department of Molecular Biology, Rocky Vista University, 8401 S Chambers Rd, Parker, CO 80134 USA
- Swedish Medical Center, 501 E. Hampden Ave. Rm 4-454, Englewood, CO 80013 USA
| |
Collapse
|
39
|
Huang Y, Wang F, Li H, Xu S, Xu W, Pan X, Hu Y, Mao L, Qian S, Pan J. Inhibition of Fibroblast Growth Factor Receptor by AZD4547 Protects Against Inflammation in Septic Mice. Inflammation 2020; 42:1957-1967. [PMID: 31321583 DOI: 10.1007/s10753-019-01056-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Sepsis is a life-threatening condition caused by the dysregulated host immune response to infection characterized by excessive secretion of inflammatory factors. AZD4547 is a selective inhibitor of fibroblast growth factor receptors that participates in the inflammatory response. The aim of this study was to investigate the inflammation-targeting effects and related molecular mechanisms of AZD4547 in sepsis using a cecal ligation and puncture model and RAW264.7 macrophages stimulated with lipopolysaccharide. AZD4547 improved the survival of CLP mice and exhibited a robust protective function against lung damage histologically. Pretreatment with AZD4547 significantly alleviated the expression of the pro-inflammatory factors IL-1β, IL-6, TNF-α, MMP9, and CXCL10 both in vivo and in vitro. In addition, AZD4547 suppressed the proliferative activity of macrophages in lung tissue and RAW264.7 macrophages. In addition, the LPS-induced phosphorylation of key proteins of NF-κB/MAPK/STAT3 pathways in RAW264.7 macrophages, such as p65, IκB-α, Erk1/2, JNK, and STAT3 proteins, could be inhibited by AZD4547 pretreatment. In conclusion, AZD4547 exerts a protective effect against excessive inflammatory damage in septic mice and may have the potential for use as an effective drug for the management of sepsis.
Collapse
Affiliation(s)
- Yueyue Huang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Fen Wang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Hao Li
- Department of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - ShunYao Xu
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Wenwei Xu
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - XiaoJun Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yufeng Hu
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Lingjie Mao
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Songzan Qian
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Jingye Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
40
|
Xu Z, Wu H, Zhang H, Bai J, Zhang Z. Interleukins 6/8 and cyclooxygenase-2 release and expressions are regulated by oxidative stress-JAK2/STAT3 signaling pathway in human bronchial epithelial cells exposed to particulate matter ≤2.5 μm. J Appl Toxicol 2020; 40:1210-1218. [PMID: 32212198 DOI: 10.1002/jat.3977] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/19/2020] [Accepted: 03/04/2020] [Indexed: 01/06/2023]
Abstract
Atmospheric particulate matter with a diameter ≤2.5 μm (PM2.5) can induce inflammation of the respiratory system, which is the pathological basis of asthma or other respiratory diseases; however, the underlying regulation mechanism has not been clearly addressed. The aim of this study was to explore the potential role of the oxidative stress-JAK/STAT signaling pathway in the inflammation of human bronchial epithelial cells induced by PM2.5. The human bronchial epithelial cell line 16HBE cells were stimulated with PM2.5 at 50 and 100 μg/mL doses for 12 or 24 hours. Intracellular reactive oxygen species (ROS) was detected using flow cytometry. Gene and protein expressions of JAK2, STAT3 and cyclooxygenase 2 (COX-2) were determined using reverse transcription-polymerase chain reaction and western blotting, respectively. The ratio of intracellular glutathione/glutathione disulfide (GSH/GSSG) and the levels of interleukin (IL)-6 and IL-8 in cellular supernatant were analyzed using enzyme-linked immunosorbent assay. The results indicated that PM2.5 treatment significantly increased gene expressions of JAK2/STAT3 and protein levels of p-JAK2/p-STAT3, accompanied by increased intracellular ROS levels, decreased GSH/GSSG ratio at 50 and 100 μg/mL of PM2.5, and significantly enhanced levels of IL-6, IL-8 and COX-2 at a dose of 100 μg/mL. Pretreatment with N-acetyl-l-cysteine (NAC) attenuated the oxidative stress induced by PM2.5; similarly, pretreatment with AG490 (an inhibitor of JAK) decreased the cytokine levels stimulated by PM2.5. Therefore, we concluded that PM2.5 exposure could activate oxidative stress-JAK2/STAT3 signaling pathway, elevate the levels of IL-6, IL-8 and COX-2 in 16HBE cells, which can be inhibited by the NAC or AG490.
Collapse
Affiliation(s)
- Zhenzhen Xu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hongyan Wu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hongmei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jianying Bai
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
41
|
Wu GC, Peng CK, Liao WI, Pao HP, Huang KL, Chu SJ. Melatonin receptor agonist protects against acute lung injury induced by ventilator through up-regulation of IL-10 production. Respir Res 2020; 21:65. [PMID: 32143642 PMCID: PMC7059294 DOI: 10.1186/s12931-020-1325-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background It is well known that ventilation with high volume or pressure may damage healthy lungs or worsen injured lungs. Melatonin has been reported to be effective in animal models of acute lung injury. Melatonin exerts its beneficial effects by acting as a direct antioxidant and via melatonin receptor activation. However, it is not clear whether melatonin receptor agonist has a protective effect in ventilator-induced lung injury (VILI). Therefore, in this study, we determined whether ramelteon (a melatonin receptor agonist) can attenuate VILI and explore the possible mechanism for protection. Methods VILI was induced by high tidal volume ventilation in a rat model. The rats were randomly allotted into the following groups: control, control+melatonin, control+ramelteon, control+luzindole, VILI, VILI+luzindole, VILI + melatonin, VILI + melatonin + luzindole (melatonin receptor antagonist), VILI + ramelteon, and VILI + ramelteon + luzindole (n = 6 per group). The role of interleukin-10 (IL-10) in the melatonin- or ramelteon-mediated protection against VILI was also investigated. Results Ramelteon treatment markedly reduced lung edema, serum malondialdehyde levels, the concentration of inflammatory cytokines in bronchoalveolar lavage fluid (BALF), NF-κB activation, iNOS levels, and apoptosis in the lung tissue. Additionally, ramelteon treatment significantly increased heat shock protein 70 expression in the lung tissue and IL-10 levels in BALF. The protective effect of ramelteon was mitigated by the administration of luzindole or an anti-IL-10 antibody. Conclusions Our results suggest that a melatonin receptor agonist has a protective effect against VILI, and its protective mechanism is based on the upregulation of IL-10 production.
Collapse
Affiliation(s)
- Geng-Chin Wu
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan. .,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Chenggong Road, Neihu, Taipei, 114, Taiwan.
| |
Collapse
|
42
|
Xinqiang Y, Quan C, Yuanyuan J, Hanmei X. Protective effect of MOTS-c on acute lung injury induced by lipopolysaccharide in mice. Int Immunopharmacol 2020; 80:106174. [PMID: 31931370 DOI: 10.1016/j.intimp.2019.106174] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/29/2019] [Accepted: 12/30/2019] [Indexed: 12/17/2022]
Abstract
MOTS-c (mitochondrial open-reading-frame of the twelve S rRNA-c), a mitochondrial-derived 16-amino acid peptide, targets the methionine-folate cycle, increases 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) levels, and eventually activates AMP-activated protein kinase (AMPK). AMPK activation can attenuate neutrophil pro-inflammatory activity and attenuates lipoteichoic acid (LTA) and lipopolysaccharide (LPS) induced acute lung injury (ALI) in mice. However, to our knowledge, the role of MOTS-c in LPS-induced ALI remains unclear. Hence, we investigated the potential effectiveness and underlying mechanism of MOTS-c against LPS-induced ALI in mice. The intraperitoneal administration of MOTS-c (5 mg/kg, i.p., bid, 6 days) before intratracheal LPS instillation attenuated body weight loss and pulmonary edema, inhibited neutrophilic tissue infiltration in lung tissue, downregulated the expression of cytokine-induced neutrophil chemoattractant-1 (CINC-1) and intercellular cell adhesion molecule-1 (ICAM-1) in lung tissues, decreased the levels of TNF-α, IL-1β, and IL-6, and increased the expression of IL-10 and SOD in serum, lung tissue, and bronchoalvelolar lavage fluid (BALF). Moreover, MOTS-c treatment significantly promoted p-AMPKα and SIRT1 expression and suppressed LPS-induced ERK, JNK, p38, p65, and STAT3 activation in the mouse lung tissues. Collectively, these findings suggest that MOTS-c plays important roles in protecting the lungs from the inflammatory effects of LPS-induced ALI. The effects of MOTS-c are probably orchestrated by activating AMPK and SIRT1, inhibiting ERK, JNK, p65, and STAT3 signaling pathways. Thus, MOTS-c appears to be a novel and promising candidate for the treatment of ALI.
Collapse
Affiliation(s)
- Yin Xinqiang
- School of Basic Medical Sciences, North Sichuan Medical College, Nanchong 673000, China; The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 211198, China
| | - Chen Quan
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 211198, China
| | - Jing Yuanyuan
- Department of Preventive Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Xu Hanmei
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 211198, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
43
|
JAK2/STAT1-mediated HMGB1 translocation increases inflammation and cell death in a ventilator-induced lung injury model. J Transl Med 2019; 99:1810-1821. [PMID: 31467427 DOI: 10.1038/s41374-019-0308-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 06/03/2019] [Accepted: 06/14/2019] [Indexed: 01/06/2023] Open
Abstract
Janus kinase 2/signal transducer and activators of transcription 1 (JAK2/STAT1) signaling is a common pathway that contributes to numerous inflammatory disorders, including different forms of acute lung injury (ALI). However, the role of JAK2/STAT1 in ventilator-induced lung injury (VILI) and its underlying mechanism remain unclear. In this study, using lipopolysaccharide (LPS) inhalation plus mechanical ventilation as VILI mouse model, we found that the administration of JAK2 inhibitor AZD1480 markedly attenuated lung destruction, diminished protein leakage, and inhibited cytokine release. In addition, when mouse macrophage-like RAW 264.7 cells were exposed to LPS and cyclic stretch (CS), AZD1480 prevented cell autophagy, reduced apoptosis, and suppressed lactate dehydrogenase release by downregulating JAK2/STAT1 phosphorylation levels and inducing HMGB1 translocation from the nucleus to the cytoplasm. Furthermore, HMGB1 and STAT1 knockdown attenuated LPS+CS-induced autophagy and apoptosis in RAW 264.7 cells. In conclusion, these findings reveal the connection between the JAK2/STAT1 pathway and HMGB1 translocation in mediating lung inflammation and cell death in VILI, suggesting that these molecules may serve as novel therapeutic targets for VILI.
Collapse
|
44
|
Hirsutanol A Attenuates Lipopolysaccharide-Mediated Matrix Metalloproteinase 9 Expression and Cytokines Production and Improves Endotoxemia-Induced Acute Sickness Behavior and Acute Lung Injury. Mar Drugs 2019; 17:md17060360. [PMID: 31213027 PMCID: PMC6627105 DOI: 10.3390/md17060360] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 12/15/2022] Open
Abstract
Activated human monocytes/macrophages, which increase the levels of matrix metalloproteinases (MMPs) and pro-inflammatory cytokines, are the essential mechanisms for the progression of sepsis. In the present study, we determined the functions and mechanisms of hirsutanolA (HA), which is isolated from the red alga-derived marine fungus Chondrostereum sp. NTOU4196, on the production of pro-inflammatory mediators produced from lipopolysaccharide (LPS)-treated THP-1 cells. Our results showed that HA suppressed LPS-triggered MMP-9-mediated gelatinolysis and expression of protein and mRNA in a concentration-dependent manner without effects on TIMP-1 activity. Also, HA significantly attenuated the levels of TNF-α, IL-6, and IL-1β from LPS-treated THP-1 cells. Moreover, HA significantly inhibited LPS-mediated STAT3 (Tyr705) phosphorylation, IκBα degradation and ERK1/2 activation in THP-1 cells. In an LPS-induced endotoxemia mouse model, studies indicated that HA pretreatment improved endotoxemia-induced acute sickness behavior, including acute motor deficits and anxiety-like behavior. HA also attenuated LPS-induced phospho-STAT3 and pro-MMP-9 activity in the hippocampus. Notably, HA reduced pathologic lung injury features, including interstitial tissue edema, infiltration of inflammatory cells and alveolar collapse. Likewise, HA suppressed the induction of phospho-STAT3 and pro-MMP-9 in lung tissues. In conclusion, our results provide pharmacological evidence that HA could be a useful agent for treating inflammatory diseases, including sepsis.
Collapse
|
45
|
Natarajan K, Meganathan V, Mitchell C, Boggaram V. Organic dust induces inflammatory gene expression in lung epithelial cells via ROS-dependent STAT-3 activation. Am J Physiol Lung Cell Mol Physiol 2019; 317:L127-L140. [PMID: 31042082 DOI: 10.1152/ajplung.00448.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure to dust in agricultural and animal environments, known as organic dust, is associated with the development of respiratory symptoms and respiratory diseases. Inflammation is a key feature of lung pathologies associated with organic dust exposure, and exposure to organic dust induces the expression of several immune and inflammatory mediators. However, information on transcription factors and cellular and molecular mechanisms controlling the production of immune and inflammatory mediators induced by organic dust is limited. In this study, we have identified STAT-3 as an important transcription factor controlling the induction of expression of immune and inflammatory mediators by poultry dust extracts in airway epithelial cells and in mouse lungs and delineated the cellular pathway for STAT-3 activation. Poultry dust extract activated STAT-3 phosphorylation in Beas2B and normal human bronchial epithelial cells and in mouse lungs. Chemical inhibition and siRNA knockdown of STAT-3 suppressed induction of immune and inflammatory mediator expression. Antioxidants suppressed the increase of STAT-3 phosphorylation induced by poultry dust extract indicating that oxidative stress [elevated reactive oxygen species (ROS) levels] is important for the activation. Chemical inhibition and siRNA knockdown experiments demonstrated that STAT-3 activation is dependent on the activation of nonreceptor tyrosine-protein kinase 2 (TYK2) and epidermal growth factor receptor (EGFR) tyrosine kinases. Our studies show that poultry dust extract controls the induction of immune and inflammatory mediator expression via a cellular pathway involving oxidative stress-mediated STAT-3 activation by TYK2 and EGFR tyrosine kinases.
Collapse
Affiliation(s)
- Kartiga Natarajan
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler , Tyler, Texas
| | - Velmurugan Meganathan
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler , Tyler, Texas
| | - Courtney Mitchell
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler , Tyler, Texas
| | - Vijay Boggaram
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler , Tyler, Texas
| |
Collapse
|
46
|
Li YZ, Chen JH, Tsai CF, Yeh WL. Anti-inflammatory Property of Imperatorin on Alveolar Macrophages and Inflammatory Lung Injury. JOURNAL OF NATURAL PRODUCTS 2019; 82:1002-1008. [PMID: 30892032 DOI: 10.1021/acs.jnatprod.9b00145] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Imperatorin is one of the furanocoumarin derivatives and exists in many medicinal herbs with anticancer, antiviral, antibacterial, and antihypertensive activities. In this study, we examined the anti-inflammatory effects of imperatorin on inflammation-associated lung diseases. Imperatorin reduced iNOS and COX-2 expression and also IL-6 and TNFα production enhanced by zymosan. Imperatorin also inhibited the signaling pathways of JAK/STAT and NF-κB. Moreover, in vivo study also revealed that zymosan-induced immune cell infiltration, pulmonary fibrosis, and edema were relieved by imperatorin in mice. We found that imperatorin exerts anti-inflammatory effects that are associated with amelioration of lung inflammation, edema, and rapid fibrosis. Studies on alveolar macrophages also reveal that imperatorin reduced the production of pro-inflammatory mediators and cytokines and inhibited pro-inflammatory JAK1/STAT3 and NF-κB signaling pathways. These results indicate that imperatorin may be a potential anti-inflammatory agent for inflammatory-associated lung diseases.
Collapse
Affiliation(s)
- Ya-Zhen Li
- Department of Biological Science and Technology , China Medical University , No. 91 Hsueh-Shih Road , Taichung , 40402 , Taiwan
| | - Jia-Hong Chen
- Department of General Surgery , Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation , Taichung , 42743 , Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology , Asia University , No. 500 Lioufeng Road , Taichung , 41354 , Taiwan
| | - Wei-Lan Yeh
- Institute of New Drug Development , China Medical University , No. 91 Hsueh-Shih Road , Taichung , 40402 , Taiwan
- Research Center for Tumor Medical Science , China Medical University , No. 91 Hsueh-Shih Road , Taichung , 40402 , Taiwan
| |
Collapse
|
47
|
LncRNA analysis of lung tissues after hUC-MSCs and FTY720 treatment of lipopolysaccharide-induced acute lung injury in mouse models. Int Immunopharmacol 2019; 71:68-75. [PMID: 30877876 DOI: 10.1016/j.intimp.2019.03.017] [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: 01/03/2019] [Revised: 02/10/2019] [Accepted: 03/06/2019] [Indexed: 11/21/2022]
Abstract
Acute lung injury (ALI), a persistent lung inflammatory response syndrome, may evolve into acute respiratory distress syndrome (ARDS). Characterized by rapid onset, critical features, and a complex etiology, ALI remains a challenging critical respiratory disease. Recently, mesenchymal stem cells (MSCs) have provided a new solution for the treatment of ALI. We built a lipopolysaccharide (LPS)-induced ALI model in mice. After treatment with human umbilical cord mesenchymal stem cells (hUC-MSCs), FTY720, or a combination of hUC-MSCs and FTY207, the lung inflammatory response was apparently attenuated. To understand the mechanism underlying MSCs treatment of ALI at the genetic level, significant differentially expressed long non-coding RNAs (lncRNAs) between the treatment and model groups were analyzed using microarray technology. Moreover, genetic gene prediction, gene ontology (GO) analysis, pathway analysis, and transcription factor (TF) prediction were carried out. The results showed that a total of 66 lncRNAs were differentially expressed in all three treatment groups, including 8 up-regulated and 58 down-regulated lncRNAs. LncRNA A_30_P01029806 and A_30_P01029194, which were down-regulated, were involved in the signaling pathways closely related to ALI. Through further TF analysis, we identified several significant TFs which lay a foundation for revealing the mechanism underlying lncRNAs treatment of ALI. LncRNA A_30_P01029806 and A_30_P01029194 may serve as candidate biomarkers in the diagnosis and treatment of ALI.
Collapse
|
48
|
Jin J, Xu W, Wan B, Wang X, Zhou Z, Miao Y, Lv T, Song Y. Topotecan Alleviates Lipopolysaccharide-Mediated Acute Lung Injury Via the NF-κB Signaling Pathway. J Surg Res 2019; 235:83-92. [DOI: 10.1016/j.jss.2018.08.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/30/2018] [Accepted: 08/24/2018] [Indexed: 12/14/2022]
|
49
|
Zheng X, Hu X, Zhang W. The phenotype of vascular smooth muscle cells co-cultured with endothelial cells is modulated by PDGFR-β/IQGAP1 signaling in LPS-induced intravascular injury. Int J Med Sci 2019; 16:1149-1156. [PMID: 31523178 PMCID: PMC6743276 DOI: 10.7150/ijms.34749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022] Open
Abstract
Background Sepsis, a leading cause of death in intensive care units, is generally associated with vascular dysfunction. However, its pathophysiological process has not been fully clarified, lacking in-depth knowledge of its pathophysiological process may hinder the improvement of diagnosis and therapy for sepsis. Hence, as the key parts of the vascular wall, the interaction between endothelial cells (ECs) and smooth muscle cells (SMCs) under septic situation need to be further studied. Methods ECs and SMCs were co-cultured using Transwell plates. Lipopolysaccharide (LPS) was used to induce sepsis. A scratch-wound assay was used to assess cell migration, and western blotting was used to assess the level of redifferentiation of SMCs as well as the expression of PDGFR-β and IQGAP1. Results Co-culture with ECs reduced the redifferentiation of SMCs induced by LPS (10 μg/ml), which was characterized by increased migration ability and decreased expression of contractile proteins (e.g., SM22 and α-SMA). The production of TNF-α could decrease the level of PDGFR-β in SMCs. Treatment of SMCs with the PDGFR-β inhibitor imatinib (5 μM) was able to counteract LPS-induced SMC redifferentiation and reduce IQGAP1 protein expression, especially when SMCs were co-cultured with ECs. Conclusion The phenotype of vascular SMCs co-cultured with ECs was modulated by IQGAP1 through the PDGFR-β pathway, which may lead to vascular remodeling and homeostasis in LPS-induced intravascular injury. This pathway could be a novel target for the treatment of vascular damage.
Collapse
Affiliation(s)
- Xia Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, 310003, P.R. China
| | - Xiaotong Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, P.R. China
| | - Wang Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, 310003, P.R. China
| |
Collapse
|
50
|
Nabavi SM, Ahmed T, Nawaz M, Devi KP, Balan DJ, Pittalà V, Argüelles-Castilla S, Testai L, Khan H, Sureda A, de Oliveira MR, Vacca RA, Xu S, Yousefi B, Curti V, Daglia M, Sobarzo-Sánchez E, Filosa R, Nabavi SF, Majidinia M, Dehpour AR, Shirooie S. Targeting STATs in neuroinflammation: The road less traveled! Pharmacol Res 2018; 141:73-84. [PMID: 30550953 DOI: 10.1016/j.phrs.2018.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/01/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022]
Abstract
JAK/STAT transduction pathway is a highly conserved pathway implicated in regulating cellular proliferation, differentiation, survival and apoptosis. Dysregulation of this pathway is involved in the onset of autoimmune, haematological, oncological, metabolic and neurological diseases. Over the last few years, the research of anti-neuroinflammatory agents has gained considerable attention. The ability to diminish the STAT-induced transcription of inflammatory genes is documented for both natural compounds (such as polyphenols) and chemical drugs. Among polyphenols, quercetin and curcumin directly inhibit STAT, while Berberis vulgaris L. and Sophora alopecuroides L extracts act indirectly. Also, the Food and Drug Administration has approved several JAK/STAT inhibitors (direct or indirect) for treating inflammatory diseases, indicating STAT can be considered as a therapeutic target for neuroinflammatory pathologies. Considering the encouraging data obtained so far, clinical trials are warranted to demonstrate the effectiveness and potential use in the clinical practice of STAT inhibitors to treat inflammation-associated neurodegenerative pathologies.
Collapse
Affiliation(s)
- Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Maheen Nawaz
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000, Pakistan
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India
| | - Devasahayam Jaya Balan
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India
| | - Valeria Pittalà
- Department of Drug Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | | | - Lara Testai
- Department of Pharmacy, University of Pisa, Pisa, via Bonanno 6 - 56126, Pisa, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress and CIBEROBN (Physiopathology of Obesity and Nutrition), University of Balearic Islands, E-07122 Palma de Mallorca, Spain.
| | - Marcos Roberto de Oliveira
- Department of Chemistry/ICET, Federal University of Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, 78060-900, Brazil
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, I-70126, Bari, Italy
| | - Suowen Xu
- University of Rochester, Aab Cardiovascular Research Institute, Rochester, NY, 14623, USA
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Valeria Curti
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maria Daglia
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Eduardo Sobarzo-Sánchez
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782, Spain; Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile
| | - Rosanna Filosa
- Consorzio Sannio Tech, Appia Str, Apollosa, BN 82030, Italy
| | - Seyed Fazel Nabavi
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran; Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Shirooie
- Department of Pharmacology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|