1
|
Kim HG, Kim JH, Yu T, Cho JY. Functional Involvement of TANK-Binding Kinase 1 in the MyD88-Dependent NF- κB Pathway Through Syk. Mediators Inflamm 2024; 2024:8634515. [PMID: 39493293 PMCID: PMC11531359 DOI: 10.1155/2024/8634515] [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: 03/31/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Inflammation is a vital immune defense mechanism regulated by Toll-like receptors (TLRs) and the nuclear factor-kappa B (NF-κB) pathway. TANK-binding kinase 1 (TBK1) is central to immunity and inflammation and influences antiviral responses and cellular processes. However, the precise role of TBK1 in modulating the NF-κB pathway through interactions with other proteins, such as spleen tyrosine kinase (Syk), remains poorly understood. As dysregulation of TBK1 and NF-κB can lead to a variety of diseases, they are important therapeutic targets. In this work, inflammatory processes involving the TBK1-Syk-NF-κB pathway were elucidated using lipopolysaccharide (LPS)-induced macrophages; human embryonic kidney 293 (HEK293) cells overexpressing MyD88, TBK1, and Syk proteins and their mutants; and real-time polymerase chain reaction (PCR), immunoblotting analyses, and kinase assays. TBK1 was activated in LPS-, poly I:C-, and Pam3CSK-stimulated macrophages. Transcript levels of TNF, NOS2, and IL1B were increased in cells overexpressing TBK1 but not in cells overexpressing TBK1 K38A. The transcription of TNF, NOS2, and IL1B and NF-κB luciferase activity were inhibited by silencing TBK1 in LPS-stimulated RAW264.7 cells and MyD88-transfected HEK293 cells. Syk was the key mediator of the TBK1-dependent NF-κB pathway and bound directly to the coiled coil domain of TBK1, which was necessary to activate Syk and the Syk-p85 pathway. This research advances the understanding of the role of TBK1 in NF-κB signaling, emphasizing Syk as a key mediator. The interaction between TBK1 and Syk has potential for precise immune modulation that can be applied to treat immune-related disorders.
Collapse
Affiliation(s)
- Han Gyung Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
2
|
Xiong W, Jia L, Cai Y, Chen Y, Gao M, Jin J, Zhu J. Evaluation of the anti-inflammatory effects of PI3Kδ/γ inhibitors for treating acute lung injury. Immunobiology 2023; 228:152753. [PMID: 37832501 DOI: 10.1016/j.imbio.2023.152753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/19/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023]
Abstract
Phosphatidylinositol 3-kinase delta (PI3Kδ) and gamma (PI3Kγ) are predominantly located in immune and hematopoietic cells. It is well-established that PI3Kδ/γ plays important roles in the immune system and participates in inflammation; hence, it could be a potential target for anti-inflammatory therapy. Currently, several PI3K inhibitors are used clinically to treat cancers with aberrant PI3K signaling; however, their role in treating acute respiratory inflammatory diseases has rarely been explored. Herein, we investigated the potential anti-inflammatory activities of several pharmacological PI3K inhibitors, including marketed drugs idelalisib (PI3Kδ), duvelisib (PI3Kδ/γ), and copanlisib (pan-PI3K with preferential α/δ) and the clinical drug eganelisib (PI3Kγ), for treating acute lung injury (ALI). In the lipopolysaccharide-induced RAW264.7 macrophage inflammatory model, the four inhibitors significantly suppressed proinflammatory cytokine expression by inhibiting the PI3K signaling pathway. Oral administration of PI3K inhibitors markedly improved lung injury in a murine model of ALI. PI3K pathway inhibition decreased inflammatory cell infiltration and totalprotein levels, as well as reduced the expression of associated lung inflammatory factors. Collectively, all four representative PI3K inhibitors exerted prominent anti-inflammatory properties, indicating that PI3K δ and/or γ inhibition could be ideal targets to treat respiratory inflammatory diseases by reducing the inflammatory response. The findings of the current study provide a new basis for utilizing PI3K inhibitors to treat acute respiratory inflammatory diseases.
Collapse
Affiliation(s)
- Wendian Xiong
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lei Jia
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Mingzhu Gao
- Department of Clinical Research Center for Jiangnan University Medical Center (Wuxi No.2 People's Hospital), Wuxi, Jiangsu 214000, China.
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
3
|
Li S, Hou J, Wang Q, Liu M, Xu X, Yang H, Li X. Angong niuhuang wan attenuates LPS-induced acute lung injury by inhibiting PIK3CG/p65/MMP9 signaling in mice based on proteomics. Heliyon 2023; 9:e20149. [PMID: 37810062 PMCID: PMC10559929 DOI: 10.1016/j.heliyon.2023.e20149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Acute lung injury (ALI) is a serious pulmonary complication that often arises from pneumonia, respiratory tract infections caused by bacteria or viruses, and other factors. It is characterized by acute onset and high mortality. Angong Niuhuang Wan (AGNHW) is a renowned emergency medicine in traditional Chinese medicine, known as the "cool open (febrile disease) three treasures" and regarded as the first of the "three treasures". Previously studies have confirmed that AGNHW has anti-inflammatory effects, improves cerebral circulation, reduces brain edema, and protects vascular endothelium. However, the active components and pharmacological mechanisms of AGNHW in treating ALI remain unclear. In this study, we confirmed that AGNHW can inhibit cytokine storm activity and reduce inflammation induced by LPS in ALI mice. We then analyzed differential proteins using proteomic technology and identified 741 differential proteins. By combining network pharmacological analysis, we deeply discussed the key active components and mechanism of AGNHW in treating ALI. By constructing the interaction network between disease and drug, we identified 21 key active components (such as Quercetin, Kaempferol, and Crocetin) and 25 potential core targets (such as PIK3CG, p65, and MMP9). These candidate targets play an important role in anti-inflammation and immune regulation. Through enrichment analysis of core targets, we found several pathways related to ALI, such as the NF-κB signaling pathway, TNF signaling pathway, and Toll-like receptor signaling pathway. This indicates that AGNHW plays a therapeutic role in ALI through multi-components, multi-targets, and multi-pathways.
Collapse
Affiliation(s)
- Sen Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jinli Hou
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mei Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xingyue Xu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
4
|
Tang L, Song D, Qi R, Zhu B, Wang X. Roles of pulmonary telocytes in airway epithelia to benefit experimental acute lung injury through production of telocyte-driven mediators and exosomes. Cell Biol Toxicol 2023; 39:451-465. [PMID: 34978009 PMCID: PMC8720540 DOI: 10.1007/s10565-021-09670-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Telocytes (TCs) are experimentally evidenced as an alternative of cell therapies for organ tissue injury and repair. The aims of the present studies are to explore direct roles of TCs and the roles of TC-derived exosomes in support of experimental acute lung injury (ALI) in vivo or in vitro. MATERIALS AND METHODS The roles of TCs in experimental ALI were firstly estimated. Phosphoinositide 3-kinase (PI3K) p110δ and α/δ/β isoform inhibitors were used in study dynamic alterations of bio-behaviors, and in expression of functional factors of TCs per se and TC-co-cultured airway epithelial cells during the activation with lipopolysaccharide (LPS). TC-driven exosomes were furthermore characterized for intercellular communication by which activated or non-activated TCs interacted with epithelia. RESULTS Our results showed that TCs mainly prevented from lung tissue edema and hemorrhage and decreased the levels of VEGF-A and MMP9 induced by LPS. Treatment with CAL101 (PI3K p110δ inhibitor) and LY294002 (PI3Kα/δ/β inhibitor) could inhibit TC movement and differentiation and increase the number of dead TCs. The expression of Mtor, Hif1α, Vegf-a, or Mmp9 mRNA increased in TCs challenged with LPS, while Mtor, Hif1α, and Vegf-a even more increased after adding CAL101 or Mtor after adding LY. The rate of epithelial cell proliferation was higher in co-culture of human bronchial epithelial (HBE) and TCs than that in HBE alone under conditions with or without LPS challenge or when cells were treated with LPS and CAL101 or LY294002. The levels of mTOR, HIF1α, or VEGF-A significantly increased in mono-cultured or co-cultured cells, challenged with LPS as compared with those with vehicle. LPS-pretreated TC-derived exosomes upregulated the expression of AKT, p-AKT, HIF1α, and VEGF-A protein of HBE. CONCLUSION The present study demonstrated that intraperitoneal administration of TCs ameliorated the severity of lung tissue edema accompanied by elevated expression of VEGF-A. TCs could nourish airway epithelial cells through nutrients produced from TCs, increasing epithelial cell proliferation, and differentiation as well as cell sensitivity to LPS challenge and PI3K p110δ and α/δ/β inhibitors, partially through exosomes released from TCs.
Collapse
Affiliation(s)
- Li Tang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital; Institute for Clinical Science Shanghai Institute of Clinical Bioinformatics Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Fudan University Shanghai Medical College, Shanghai, China
| | - Dongli Song
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital; Institute for Clinical Science Shanghai Institute of Clinical Bioinformatics Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Fudan University Shanghai Medical College, Shanghai, China.
| | - Ruixue Qi
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital; Institute for Clinical Science Shanghai Institute of Clinical Bioinformatics Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Fudan University Shanghai Medical College, Shanghai, China
| | - Bijun Zhu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital; Institute for Clinical Science Shanghai Institute of Clinical Bioinformatics Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital; Institute for Clinical Science Shanghai Institute of Clinical Bioinformatics Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases Jinshan Hospital Centre for Tumor Diagnosis and Therapy, Fudan University Shanghai Medical College, Shanghai, China.
| |
Collapse
|
5
|
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: 16] [Impact Index Per Article: 5.3] [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
|
6
|
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: 0.3] [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
|
7
|
Yao L, Tang Y, Chen J, Li J, Wang H, Lu M, Gao L, Liu F, Chang P, Liu X, Tang H. Impaired airway epithelial barrier integrity was mediated by PI3Kδ in a mouse model of lipopolysaccharide-induced acute lung injury. Int Immunopharmacol 2021; 95:107570. [PMID: 33773208 DOI: 10.1016/j.intimp.2021.107570] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 01/03/2023]
Abstract
Cell-cell junctions are critical for the maintenance of cellular as well as tissue polarity and integrity. Dysfunction of airway epithelial barrier has been shown to be involved in the pathogenesis of acute lung injury (ALI). Yet the role of phosphatidylinositol 3-kinase delta (PI3Kδ) in dysregulation of airway epithelial barrier integrity in ALI has not been addressed. Mice were subjected to intratracheal instillation of lipopolysaccharide (LPS) to generate a ALI model. Two pharmacological inhibitors of PI3Kδ, IC87114 and AMG319, were respectively given to the mice. Expression of p110δ and its downstream substrate phospho-AKT (Ser473) was increased in LPS-exposed lungs. These increases were inhibited by IC87114 or AMG319. LPS led to pronounced lung injury that was accompanied by significant airway neutrophil recruitment and bronchial epithelial morphological alterations 72 h after exposure. We also found compromised expression of adherens junction protein E-cadherin and tight junction protein claudin-2 in the airway epithelial cells. Treatment with either IC87114 or AMG319 not only attenuated LPS-induced edema, lung injury and neutrophilc inflammation, reduced total protein concentration and IL-6, TNF-α secretion in BALF, but also restored epithelial E-cadherin and claudin-2 expression. In summary, our results showed that LPS can induce a delayed effect on airway epithelial barrier integrity that is mediated by PI3Kδ in a mouse model of ALI.
Collapse
Affiliation(s)
- Lihong Yao
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ying Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiahui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Wang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Lu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lijuan Gao
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xingxing Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Haixiong Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Pereira CMC, Júnior GJD, Lima JVDN, Alves Lemos SI, da Rocha Rodrigues L, Dos Santos Ferreira J, Araújo ASML, de Oliveira JC, Monteiro CE, Franco ÁX, Pereira Alves EH, Oliveira Silva FG, de Carvalho Filgueiras M, Soares PMG, Pereira Vasconcelos DF, de Oliveira JS, de Brito TV, Barbosa ALR. Phosphatidylinositol 3-kinase gamma participates in nimesulide-induced hepatic damage. J Pharm Pharmacol 2021; 73:496-504. [PMID: 33793830 DOI: 10.1093/jpp/rgaa049] [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: 07/28/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To evaluate the participation of the phosphatidylinositol 3-kinase pathway in the liver damage caused by nimesulide. METHODS Liver damage been induced by nimesulide. Mice were treated with either 2% dimethyl sulfoxide or AS605240, a phosphatidylinositol 3-kinase gamma pathway antagonist. Blood samples were collected for function assays of liver. The liver was removed for analysis of liver weight/animal weight ratio, histopathological parameters, oxidative and nitrous stress, cytokine levels, and the immunostaining for cyclooxygenase 2 and nuclear factor kappa B. KEY FINDINGS Liver injured by nimesulide and treated with phosphatidylinositol 3-kinase gamma inhibitor significantly reversed (P < 0.05) the damage; it decreased the liver weight/animal weight ratio, histopathological scores, and neutrophil infiltration, consequently reducing oxidative stress. In addition, we show that phosphatidylinositol 3-kinase gamma is associated with hepatic damage induced by nimesulide, because it altered liver function and increased the protein immunostaining of cyclooxygenase 2 and nuclear factor kappa B in the liver tissue of nimesulide-treated animals. CONCLUSIONS The findings from the present study allows us to infer that nimesulide causes liver damage through the phosphatidylinositol 3-kinase gamma pathway.
Collapse
Affiliation(s)
- Cynthia Maria C Pereira
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Genilson José Dias Júnior
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - José Victor do N Lima
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Sarah Izabelly Alves Lemos
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Lauanda da Rocha Rodrigues
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Jayro Dos Santos Ferreira
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Anna Sofia Miranda Loiola Araújo
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Joveline Costa de Oliveira
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Carlos Eduardo Monteiro
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Álvaro Xavier Franco
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Even Herlany Pereira Alves
- Laboratory of Analysis and Histological Processing (LAPHIS), Department of Biomedicine, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Francisca Géssica Oliveira Silva
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Marcelo de Carvalho Filgueiras
- Laboratory of Muscle Morphology and Physiology (LAMFIM), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Pedro M G Soares
- Laboratory of Physiopharmacology Study of Gastrointestinal Tract (LEFFAG), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Daniel Fernando Pereira Vasconcelos
- Laboratory of Analysis and Histological Processing (LAPHIS), Department of Biomedicine, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Jefferson Soares de Oliveira
- Biochemistry Laboratory of Laticifers Plants (LABPL), Department of Biomedicine, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - Tarcisio Vieira de Brito
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| | - André Luiz Reis Barbosa
- Laboratory of Experimental Physiopharmacology (LAFFEX), Department of Physioterapy, Federal University of the Parnaíba Delta, Parnaíba, Piauí, Brazil
| |
Collapse
|
9
|
Zheng F, Zhu J, Zhang W, Fu Y, Lin Z. Thal protects against paraquat-induced lung injury through a microRNA-141/HDAC6/IκBα-NF-κB axis in rat and cell models. Basic Clin Pharmacol Toxicol 2021; 128:334-347. [PMID: 33015978 PMCID: PMC7894280 DOI: 10.1111/bcpt.13505] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022]
Abstract
The protective functions of thalidomide in paraquat (PQ)-induced injury have been reported. But the mechanisms remain largely unknown. In this research, a PQ-treated rat model was established and further treated with thalidomide. Oedema and pathological changes, oxidative stress, inflammation, fibrosis and cell apoptosis in rat lungs were detected. A PQ-treated RLE-6TN cell model was constructed, and the viability and apoptosis rate of cells were measured. Differentially expressed microRNAs (miRNAs) after thalidomide administration were screened out. Binding relationship between miR-141 and histone deacetylase 6 (HDAC6) was validated. Altered expression of miR-141 and HDAC6 was introduced to identify their involvements in thalidomide-mediated events. Consequently, thalidomide administration alone exerted no damage to rat lungs; in addition it reduced PQ-induced oedema. The oxidative stress, inflammation and cell apoptosis in rat lungs were reduced by thalidomide. In RLE-6TN cells, thalidomide increased cell viability and decreased apoptosis. miR-141 was responsible for thalidomide-mediated protective events by targeting HDAC6. Overexpression of HDAC6 blocked the protection of thalidomide against PQ-induced injury via activating the IkBα-NF-κB signalling pathway. Collectively, this study evidenced that thalidomide protects lung tissues from PQ-induced injury through a miR-141/HDAC6/IkBα-NF-κB axis.
Collapse
Affiliation(s)
- Fenshuang Zheng
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Junbo Zhu
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Wei Zhang
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Yangshan Fu
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Zhaoheng Lin
- Department of Critical Care MedicinePeople's Hospital of Xishuangbanna Dai Nationality Autonomous PrefecturePingpongChina
| |
Collapse
|
10
|
Gao Z, Sui J, Fan R, Qu W, Dong X, Sun D. Emodin Protects Against Acute Pancreatitis-Associated Lung Injury by Inhibiting NLPR3 Inflammasome Activation via Nrf2/HO-1 Signaling. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1971-1982. [PMID: 32546964 PMCID: PMC7247729 DOI: 10.2147/dddt.s247103] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022]
Abstract
Aim Lung injury is a common complication of acute pancreatitis (AP), which leads to the development of acute respiratory distress syndrome and causes high mortality. In the present study, we investigated the therapeutic effect of emodin on AP-induced lung injury and explored the molecular mechanisms involved. Materials and Methods Thirty male Sprague-Dawley rats were randomly divided into AP (n=24) and normal (n=6) groups. Rats in the AP group received a retrograde injection of 5% sodium taurocholate into the biliary-pancreatic duct and then randomly assigned to untreated, emodin, combined emodin and ML385, and dexamethasone (DEX) groups. Pancreatic and pulmonary injury was assessed using H&E staining. In in vitro study, rat alveolar epithelial cell line L2 cells were exposed to lipopolysaccharide and treated with emodin. Nrf2 siRNA pool was applied for the knockdown of Nrf2. The contents of the pro-inflammatory cytokines in the bronchoalveolar lavage fluid and lung were determined using enzyme-linked immunosorbent assay. The expressions of related mRNAs and proteins in the lung or L2 cells were detected using real-time polymerase chain reaction, Western blot, immunohistochemistry and immunofluorescence. Key Findings Emodin administration alleviated pancreatic and pulmonary injury of rats with AP. Emodin administration suppressed the production of proinflammatory cytokines, downregulated NLRP3, ASC and caspase-1 expressions and inhibited NF-κB nuclear accumulation in the lung. In addition, Emodin increased Nrf2 nuclear translocation and upregulated HO-1 expression. Moreover, the anti-inflammatory effect of emodin was blocked by Nrf2 inhibitor ML385. Conclusion Emodin effectively protects rats against AP-associated lung injury by inhibiting NLRP3 inflammasome activation via Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Zhenming Gao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Jidong Sui
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Rong Fan
- Department of International Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Weikun Qu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Xuepeng Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| | - Deguang Sun
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116027, People's Republic of China
| |
Collapse
|
11
|
Kim J, Jung KH, Yoo J, Park JH, Yan HH, Fang Z, Lim JH, Kwon SR, Kim MK, Park HJ, Hong SS. PBT-6, a Novel PI3KC2γ Inhibitor in Rheumatoid Arthritis. Biomol Ther (Seoul) 2020; 28:172-183. [PMID: 31739383 PMCID: PMC7059814 DOI: 10.4062/biomolther.2019.153] [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] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/29/2022] Open
Abstract
Phosphoinositide 3-kinase (PI3K) is considered as a promising therapeutic target for rheumatoid arthritis (RA) because of its involvement in inflammatory processes. However, limited studies have reported the involvement of PI3KC2γ in RA, and the underlying mechanism remains largely unknown. Therefore, we investigated the role of PI3KC2γ as a novel therapeutic target for RA and the effect of its selective inhibitor, PBT-6. In this study, we observed that PI3KC2γ was markedly increased in the synovial fluid and tissue as well as the PBMCs of patients with RA. PBT-6, a novel PI3KC2γ inhibitor, decreased the cell growth of TNF-mediated synovial fibroblasts and LPS-mediated macrophages. Furthermore, PBT-6 inhibited the PI3KC2γ expression and PI3K/ AKT signaling pathway in both synovial fibroblasts and macrophages. In addition, PBT-6 suppressed macrophage migration via CCL2 and osteoclastogenesis. In CIA mice, it significantly inhibited the progression and development of RA by decreasing arthritis scores and paw swelling. Three-dimensional micro-computed tomography confirmed that PBT-6 enhanced the joint structures in CIA mice. Taken together, our findings suggest that PI3KC2γ is a therapeutic target for RA, and PBT-6 could be developed as a novel PI3KC2γ inhibitor to target inflammatory diseases including RA.
Collapse
Affiliation(s)
- Juyoung Kim
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Kyung Hee Jung
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Jaeho Yoo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jung Hee Park
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Hong Hua Yan
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Zhenghuan Fang
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Joo Han Lim
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Seong-Ryul Kwon
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Myung Ku Kim
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Soon-Sun Hong
- Department of Medicine, College of Medicine, Inha University, Incheon 22313, Republic of Korea
| |
Collapse
|
12
|
Jin R, Xiao AY, Li J, Wang M, Li G. PI3Kγ (Phosphoinositide 3-Kinase-γ) Inhibition Attenuates Tissue-Type Plasminogen Activator-Induced Brain Hemorrhage and Improves Microvascular Patency After Embolic Stroke. Hypertension 2019; 73:206-216. [PMID: 30571560 DOI: 10.1161/hypertensionaha.118.12001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Genetic and pharmacological inhibition of the PI3Kγ (phosphoinositide 3-kinase-γ) exerts anti-inflammatory and protective effects in a number of inflammatory and autoimmune diseases. SHRs (spontaneously hypertensive rats) subjected to embolic middle cerebral occlusion were treated with AS605240 (30 mg/kg) at 2 or 4 hours, tPA (tissue-type plasminogen activator; 10 mg/kg) at 2 or 6 hours, or AS605240 at 4 hours plus tPA at 6 hours. Infarct volume, brain hemorrhage, neurological function, microvascular thrombosis, and cerebral microvessel patency were examined. We found that treatment with AS605240 alone at 2 hours or the combination treatment with AS605240 at 4 hours and tPA at 6 hours significantly reduced infarct volume and neurological deficits at 3 days after stroke compared with ischemic rats treated with saline, AS605240 alone at 4 hours, and tPA alone at 6 hours. Moreover, the combination treatment effectively prevented the delayed tPA-induced cerebral hemorrhage. These protective effects are associated with reduced disruption of the blood-brain barrier, reduced downstream microvascular thrombosis, and improved microvascular patency by AS605240. Inhibition of the NF-κB (nuclear transcription factor-κB)-dependent MMP (matrix metalloproteinase)-9 and PAI-1 (plasminogen activator inhibitor-1) in the ischemic brain endothelium may underlie the neurovascular protective effect of AS605240. In addition, the combination treatment significantly reduced circulating platelet P-selectin expression and platelet-leukocyte aggregation compared with ischemic rats treated with saline or tPA alone at 6 hours. In conclusion, inhibition of PI3Kγ with AS605240 reduces delayed tPA-induced intracerebral hemorrhage and improves microvascular patency, which likely contributes to neuroprotective effect of the combination treatment.
Collapse
Affiliation(s)
- Rong Jin
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA (R.J., M.W., G.L.)
| | - Adam Y Xiao
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport (A.Y.X.)
| | | | - Min Wang
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA (R.J., M.W., G.L.)
| | - Guohong Li
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA (R.J., M.W., G.L.)
| |
Collapse
|
13
|
Xie Y, He Q, Chen H, Lin Z, Xu Y, Yang C. Crocin ameliorates chronic obstructive pulmonary disease-induced depression via PI3K/Akt mediated suppression of inflammation. Eur J Pharmacol 2019; 862:172640. [PMID: 31491407 DOI: 10.1016/j.ejphar.2019.172640] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 01/19/2023]
Abstract
Clinical studies have indicated the co-occurrence of chronic obstructive pulmonary disease (COPD) and psychiatric disorders, for example, comorbid depression. However, the underlying mechanism is rarely addressed. The present study aimed to investigate the mechanism of COPD-induced depression and the psychological and physiological effects of crocin, an active constituent of Crocus sativus L. C57BL/6 mice were randomly exposed to cigarette smoke for 7 weeks to establish COPD animal model. Crocin (50 mg/kg), Dexamethasone (2 mg/kg) and IGF-1 (2 mg/kg) were respectively injected to mice once a day. The FEV1/FVC ratio and the mean alveolus area of lung tissue demonstrated the COPD model was successfully established by cigarette smoke. Crocin administration significantly reversed markers of depression [loss of body weight, sucrose preference, and elevation of immobile time in tail-suspension tests (TST) and in forced swimming tests (FST)]. Besides, crocin treatment significantly inhibited the numbers of inflammatory cells (macrophages, neutrophils, and lymphocytes), suppressed the infiltration of peribronchial inflammatory cells, and reduced the concentration of proinflammatory cytokines in bronchoalveolar lavage (BAL) fluid and lung tissue. Crocin also reduced proinflammatory cytokines in the hippocampus. In exploring associated mechanisms, we discovered that crocin blunted cigarette smoke-induced IκB phosphorylation and degradation, and NF-κBp65 nuclear translocation. IGF-1, an activator of PI3K, abrogated the effect of crocin against cigarette smoke-induced activation of the NF-κB pathway. Together, these results showed that an inflammatory mechanism might be involved in the pathogenesis of COPD with comorbid depression. Crocin exhibited significant effects through the regulation of PI3K/Akt-mediated inflammatory pathways.
Collapse
Affiliation(s)
- Yupeng Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Qiuxiang He
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hong Chen
- Department of Psychiatry, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zijiang Lin
- Department of Psychiatry, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi Xu
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Chuang Yang
- Department of Psychiatry, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
14
|
Zheng X, Zhang W, Wang Z. Simvastatin preparations promote PDGF-BB secretion to repair LPS-induced endothelial injury through the PDGFRβ/PI3K/Akt/IQGAP1 signalling pathway. J Cell Mol Med 2019; 23:8314-8327. [PMID: 31576676 PMCID: PMC6850957 DOI: 10.1111/jcmm.14709] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/14/2019] [Accepted: 08/29/2019] [Indexed: 01/11/2023] Open
Abstract
Endothelial barrier dysfunction is a critical pathophysiological process of sepsis. Impaired endothelial cell migration is one of the main reasons for endothelial dysfunction. Statins may have a protective effect on endothelial barrier function. However, the effect and mechanism of statins on lipopolysaccharide (LPS)‐induced endothelial barrier dysfunction remain unclear. Simvastatin (SV) was loaded in nanostructured lipid carriers to produce SV nanoparticles (SV‐NPs). Normal SV and SV‐NPs were used to treat human umbilical vein vascular endothelial cells (HUVECs) injured by LPS. Barrier function was evaluated by monitoring cell monolayer permeability and transendothelial electrical resistance, and cell migration ability was measured by a wound healing assay. LY294002 and imatinib were used to inhibit the activity of PI3K/Akt and platelet‐derived growth factor receptor (PDGFR) β. IQ‐GTPase‐activating protein 1 (IQGAP1) siRNA was used to knockdown endogenous IQGAP1, which was used to verify the role of the PDGFRβ/PI3K/Akt/IQGAP1 pathway in SV/SV‐NPs‐mediated barrier protection in HUVECs injured by LPS. The results show that SV/SV‐NPs promoted the migration and decreased the permeability of HUVECs treated with LPS, and the efficacy of the SV‐NPs exceeded that of SV significantly. LY294002, imatinib and IQGAP1 siRNA all suppressed the barrier protection of SV/SV‐NPs. SV/SV‐NPs promoted the secretion of platelet‐derived growth factor‐BB (PDGF‐BB) and activated the PDGFRβ/PI3K/Akt/IQGAP1 pathway. SV preparations restored endothelial barrier function by restoring endothelial cell migration, which is involved in the regulation of the PDGFRβ/PI3K/Akt/IQGAP1 pathway and PDGF‐BB secretion. As an appropriate formulation for restoring endothelial dysfunction, SV‐NPs may be more effective than SV.
Collapse
Affiliation(s)
- Xia Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wang Zhang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhen Wang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Shang S, Liu L, Wu X, Fan F, Hu E, Wang L, Ding Y, Zhang Y, Lu X. Inhibition of PI3Kγ by AS605240 Protects tMCAO Mice by Attenuating Pro-Inflammatory Signaling and Cytokine Release in Reactive Astrocytes. Neuroscience 2019; 415:107-120. [PMID: 31195053 DOI: 10.1016/j.neuroscience.2019.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 05/31/2019] [Accepted: 06/02/2019] [Indexed: 12/29/2022]
Abstract
The intense and prolonged inflammatory response after ischemic stroke significantly contributes to the secondary neural injury. PI3Kγ, which is involved in the regulation of vascular permeability, chemotactic leukocyte migration and microglia activation, is a key target for intervention in the inflammatory response. In this study, we identified the protective effect of the PI3Kγ inhibitor AS605240 against stroke-related injury in the mouse model of transient intraluminal middle cerebral artery occlusion (tMCAO). The results showed that administration of AS605240 could improve the neurological function score, reduce the infarct size and decrease astrocyte activation in the tMCAO mice after injury. The inhibitory effect of AS605240 on microglia activation is relatively clear. Therefore, in this study, the effects of AS605240 on astrocytes were studied in cell cultures. IL-6 and its soluble receptor were used to construct the astrocyte activation model. AS605240 treatment significantly reduced the astrocyte activation markers and the morphological changes of cells. We also identified 13 inflammatory factors whose expression was significantly upregulated by IL-6/sIL-6R and significantly inhibited by AS605240 at the protein level, and seven of those factors were verified at the mRNA level. These results indicated that specific inhibition of PI3Kγ could reduce astrocyte activation induced by inflammation, which might aid the repair and remodeling of neurons in the later stage after ischemic stroke.
Collapse
Affiliation(s)
- Sen Shang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Lin Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Xingjuan Wu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Fan Fan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Erling Hu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Leilei Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Yan Ding
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Yali Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China
| | - Xiaoyun Lu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, PR China.
| |
Collapse
|
16
|
Different concentrations of lipopolysaccharide regulate barrier function through the PI3K/Akt signalling pathway in human pulmonary microvascular endothelial cells. Sci Rep 2018; 8:9963. [PMID: 29967433 PMCID: PMC6028427 DOI: 10.1038/s41598-018-28089-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
Lipopolysaccharide (LPS) can lead to vascular endothelial barrier dysfunction, which often results in acute lung injury and acute respiratory distress syndrome. However, the effects of different concentrations of LPS on human pulmonary microvascular endothelial barrier function and the involvement of the phosphatidylinositol-3-kinase-serine/threonine kinase (PI3K/Akt) pathway in this process remain unclear. Human pulmonary microvascular endothelial cells (HPMECs) were stimulated with different doses of LPS, and barrier function was examined by determining cell monolayer permeability, cell migration, and the expression of intercellular junction proteins (VE-Cadherin, Claudin-5, and Connexin-43). LY294002 was used to inhibit PI3K to verify the role of the PI3K/Akt pathway in the regulation of barrier function in HPMECs stimulated by LPS. Low doses of LPS increased HPMEC migration, up-regulated VE-Cadherin and Claudin-5 expression, down-regulated Connexin-43 expression, and promoted Akt phosphorylation, which could collectively decrease monolayer permeability. In contrast, high doses of LPS suppressed HPMEC migration, down-regulated the expression of VE-Cadherin and Claudin-5, up-regulated Connexin-43 expression, and reduced Akt phosphorylation, which could collectively increase monolayer permeability. LPS has a biphasic effect on HPMEC barrier function through the PI3K/Akt pathway, and this effect is concentration-dependent.
Collapse
|
17
|
Teng J, Liu M, Su Y, Li K, Sui N, Wang S, Li L, Sun Y, Wang Y. Down-regulation of GRP78 alleviates lipopolysaccharide-induced acute kidney injury. Int Urol Nephrol 2018; 50:2099-2107. [PMID: 29915879 DOI: 10.1007/s11255-018-1911-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 06/07/2018] [Indexed: 10/14/2022]
Abstract
PURPOSE Acute kidney injury (AKI) is accompanied with life-threatening sepsis. It is necessary to develop effective therapy agent or strategy for treating AKI. LPS is a primary pathogenic factor that induces sepsis. Glucose-regulated protein 78 (GRP78) is closely related to cell injuries. The objective of this study was to examine the role of GRP78 in LPS-induced AKI. METHODS Cell counting kit-8 (CCK-8) assay and flow cytometry (FCM) were respectively performed to assess the cell viability and apoptosis. Available commercial kits were used to detect the reactive oxygen species (ROS) contents and the activity of oxidative indicators. The expressions of the relevant factors were determined by real-time PCR (RT-PCR) and Western blot. RESULTS The results showed that the expression of GRP78 was apparently increased by LPS treatment, and that the down-regulation of GRP78 by small RNA interference improved the proliferation ability of renal cells in comparison to LPS group. The LPS-induced immune response and oxidative stress was alleviated by the depletion of GRP78. Moreover, the LPS-induced apoptosis was reduced in the GRP78 group by regulating the expression of mitochondrial apoptosis (Bcl-2, Bax) and endoplasmic reticulum (ER) stress (CHOP, caspase-12)-associated proteins. In addition, the protective role of GRP78 reduction was partly related to the balance of NF-κB/IκB. CONCLUSIONS Down-regulation of GRP78 attenuated LPS-induced AKI through inhibiting immune response/oxidative stress-associated apoptosis.
Collapse
Affiliation(s)
- Jinlong Teng
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingjun Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuan Su
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kun Li
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Na Sui
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shibo Wang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liandi Li
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yunbo Sun
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yongbin Wang
- Department of Emergency Medicine, The Affiliated hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
| |
Collapse
|
18
|
Inhibition of Phosphatidylinositol 3-kinease suppresses formation and progression of experimental abdominal aortic aneurysms. Sci Rep 2017; 7:15208. [PMID: 29123158 PMCID: PMC5680315 DOI: 10.1038/s41598-017-15207-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/23/2017] [Indexed: 01/10/2023] Open
Abstract
Accumulating evidence suggests an important role of Phosphatidylinositol 3-kinease (PI3K) pathway in inflammatory cells infiltration. Given the essential role of inflammatory cells infiltration during the formation and progression of abdominal aortic aneurysm (AAA), to investigate the possibility of preventing AAA formation and progression via targeting PI3K is anticipated. Here, experimental AAAs was created in rats by transient intraluminal porcine pancreatic elastase (PPE) infusion into the infrarenal aorta firstly. AAAs rats were administrated with vehicle or Wortmannin during the period of day 0 to day 28 after PPE infusion. The aortic diameter of rats treated with Wortmannin was significantly smaller than those treated with vehicle. Meanwhile, Elastin destruction score and SMC destruction score were significantly decreased in rats treated with Wortmannin. Furthermore, histological analysis revealed infiltration of inflammatory cells were significantly reduced in rats treated with Wortmannin. Finally, the mRNA expression of PI3K and protein expression of pAKT in human abdominal aneurismal aorta tissues was elevated as compare to normal aorta. Our study revealed that PI3K inhibitor suppresses experimental AAAs formation and progression, through mechanisms likely related to impairing inflammation cells infiltration and median elastin degradation. These findings indicated that PI3K inhibitor may hold substantial translation value for AAA diseases.
Collapse
|
19
|
Liu W, Zhu H, Fang H. Propofol Potentiates Sevoflurane-Induced Inhibition of Nuclear Factor--κB-Mediated Inflammatory Responses and Regulation of Mitogen-Activated Protein Kinases Pathways via Toll-like Receptor 4 Signaling in Lipopolysaccharide-Induced Acute Lung Injury in Mice. Am J Med Sci 2017; 354:493-505. [DOI: 10.1016/j.amjms.2017.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 06/01/2017] [Accepted: 06/19/2017] [Indexed: 12/24/2022]
|
20
|
Fan TT, Feng XY, Yang YZ, Gao F, Liu Q. Downregulation of PI3K-γ in a mouse model of sepsis-induced myocardial dysfunction. Cytokine 2017; 96:208-216. [PMID: 28458167 DOI: 10.1016/j.cyto.2017.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 04/17/2017] [Accepted: 04/18/2017] [Indexed: 01/25/2023]
Abstract
A key component during sepsis is the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway, of which the PI3K-γ isoform is a major regulator in many inflammatory responses. However, the role of PI3K-γ in the development of sepsis-induced myocardial dysfunction (SIMD) is unknown. In this study, we established a model of SIMD induced by lipopolysaccharide (LPS), subsequently used the selective inhibitor LY294002 and AS605240 to block the effect of PI3K and PI3K-γ, respectively. Cardiac function was evaluated by echocardiography, hearts were obtained for histological and protein expression examinations. ELISA was used to measure the serum levels of tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), cardiac troponin I (cTnI) and heart-type fatty acid binding protein (H-FABP). LPS-treated mice showed an increase to cardiac inflammation, myocardial damage and production of TNF-α, IL-6, NF-κB, cTnI and H-FABP. Administration of AS605240 to LPS-treated mice reduced some patho-physiological characteristics of SIMD and reduced TNF-α, IL-6, cTnI and H-FABP production. However, administration of LY294002 did not improve those same conditions. The results showed that PI3K-γ is likely a crucial element in SIMD by regulating the PI3K/Akt pathway, and become a new marker of myocardial injury. Inhibition of PI3K-γ might be a potential therapeutic target in SIMD.
Collapse
Affiliation(s)
- Ting-Ting Fan
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China; Department of Emergency, Chengdu First People's Hospital, Sichuan 610016, PR China
| | - Xuan-Yun Feng
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yuan-Zheng Yang
- Department of Critical Care Medicine, The Affiliated Hospital of Hainan Medical College, Hainan 571101, PR China
| | - Feng Gao
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Qiong Liu
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
21
|
Juss JK, House D, Amour A, Begg M, Herre J, Storisteanu DML, Hoenderdos K, Bradley G, Lennon M, Summers C, Hessel EM, Condliffe A, Chilvers ER. Acute Respiratory Distress Syndrome Neutrophils Have a Distinct Phenotype and Are Resistant to Phosphoinositide 3-Kinase Inhibition. Am J Respir Crit Care Med 2016; 194:961-973. [PMID: 27064380 PMCID: PMC5067816 DOI: 10.1164/rccm.201509-1818oc] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/29/2016] [Indexed: 01/08/2023] Open
Abstract
RATIONALE Acute respiratory distress syndrome is refractory to pharmacological intervention. Inappropriate activation of alveolar neutrophils is believed to underpin this disease's complex pathophysiology, yet these cells have been little studied. OBJECTIVES To examine the functional and transcriptional profiles of patient blood and alveolar neutrophils compared with healthy volunteer cells, and to define their sensitivity to phosphoinositide 3-kinase inhibition. METHODS Twenty-three ventilated patients underwent bronchoalveolar lavage. Alveolar and blood neutrophil apoptosis, phagocytosis, and adhesion molecules were quantified by flow cytometry, and oxidase responses were quantified by chemiluminescence. Cytokine and transcriptional profiling were used in multiplex and GeneChip arrays. MEASUREMENTS AND MAIN RESULTS Patient blood and alveolar neutrophils were distinct from healthy circulating cells, with increased CD11b and reduced CD62L expression, delayed constitutive apoptosis, and primed oxidase responses. Incubating control cells with disease bronchoalveolar lavage recapitulated the aberrant functional phenotype, and this could be reversed by phosphoinositide 3-kinase inhibitors. In contrast, the prosurvival phenotype of patient cells was resistant to phosphoinositide 3-kinase inhibition. RNA transcriptomic analysis revealed modified immune, cytoskeletal, and cell death pathways in patient cells, aligning closely to sepsis and burns datasets but not to phosphoinositide 3-kinase signatures. CONCLUSIONS Acute respiratory distress syndrome blood and alveolar neutrophils display a distinct primed prosurvival profile and transcriptional signature. The enhanced respiratory burst was phosphoinositide 3-kinase-dependent but delayed apoptosis and the altered transcriptional profile were not. These unexpected findings cast doubt over the utility of phosphoinositide 3-kinase inhibition in acute respiratory distress syndrome and highlight the importance of evaluating novel therapeutic strategies in patient-derived cells.
Collapse
Affiliation(s)
- Jatinder K. Juss
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - David House
- Refractory Respiratory Inflammation Discovery Performance Unit and
| | - Augustin Amour
- Refractory Respiratory Inflammation Discovery Performance Unit and
| | - Malcolm Begg
- Refractory Respiratory Inflammation Discovery Performance Unit and
| | - Jurgen Herre
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | - Kim Hoenderdos
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Glyn Bradley
- Target Sciences, GlaxoSmithKline, Stevenage, United Kingdom; and
| | - Mark Lennon
- Target Sciences, GlaxoSmithKline, Stevenage, United Kingdom; and
| | - Charlotte Summers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Edith M. Hessel
- Refractory Respiratory Inflammation Discovery Performance Unit and
| | - Alison Condliffe
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Edwin R. Chilvers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
22
|
Huang X, Dai Z, Cai L, Sun K, Cho J, Albertine KH, Malik AB, Schraufnagel DE, Zhao YY. Endothelial p110γPI3K Mediates Endothelial Regeneration and Vascular Repair After Inflammatory Vascular Injury. Circulation 2016; 133:1093-103. [PMID: 26839042 DOI: 10.1161/circulationaha.115.020918] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 01/29/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND The integrity of endothelial monolayer is a sine qua non for vascular homeostasis and maintenance of tissue-fluid balance. However, little is known about the signaling pathways regulating regeneration of the endothelial barrier after inflammatory vascular injury. METHODS AND RESULTS Using genetic and pharmacological approaches, we demonstrated that endothelial regeneration selectively requires activation of p110γPI3K signaling, which thereby mediates the expression of the endothelial reparative transcription factor Forkhead box M1 (FoxM1). We observed that FoxM1 induction in the pulmonary vasculature was inhibited in mice treated with a p110γ-selective inhibitor and in Pik3cg(-/-) mice after lipopolysaccharide challenge. Pik3cg(-/-) mice exhibited persistent lung inflammation induced by sepsis and sustained increase in vascular permeability. Restoration of expression of either p110γ or FoxM1 in pulmonary endothelial cells of Pik3cg(-/-) mice restored endothelial regeneration and normalized the defective vascular repair program. We also observed diminished expression of p110γ in pulmonary vascular endothelial cells of patients with acute respiratory distress syndrome, suggesting that impaired p110γ-FoxM1 vascular repair signaling pathway is a critical factor in persistent leaky lung microvessels and edema formation in the disease. CONCLUSIONS We identify p110γ as the critical mediator of endothelial regeneration and vascular repair after sepsis-induced inflammatory injury. Thus, activation of p110γ-FoxM1 endothelial regeneration may represent a novel strategy for the treatment of inflammatory vascular diseases.
Collapse
Affiliation(s)
- Xiaojia Huang
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Zhiyu Dai
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Lei Cai
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Kai Sun
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Jaehyung Cho
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Kurt H Albertine
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Asrar B Malik
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Dean E Schraufnagel
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - You-Yang Zhao
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.).
| |
Collapse
|
23
|
Bueno-Silva B, Kawamoto D, Ando-Suguimoto ES, Alencar SM, Rosalen PL, Mayer MPA. Brazilian Red Propolis Attenuates Inflammatory Signaling Cascade in LPS-Activated Macrophages. PLoS One 2015; 10:e0144954. [PMID: 26660901 PMCID: PMC4684384 DOI: 10.1371/journal.pone.0144954] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/27/2015] [Indexed: 12/16/2022] Open
Abstract
Although previous studies suggested an anti-inflammatory property of Brazilian red propolis (BRP), the mechanisms involved in the anti-inflammatory effects of BRP and its activity on macrophages were still not elucidated. This study aimed to evaluate whether BRP attenuates the inflammatory effect of LPS on macrophages and to investigate its underlying mechanisms. BRP was added to RAW 264.7 murine macrophages after activation with LPS. NO production, cell viability, cytokines profile were evaluated. Activation of inflammatory signaling pathways and macrophage polarization were determined by RT-qPCR and Western blot. BRP at 50 μg/ml inhibited NO production by 78% without affecting cell viability. Cd80 and Cd86 were upregulated whereas mrc1 was down regulated by BRP indicating macrophage polarization at M1. BRP attenuated the production of pro-inflammatory mediators IL-12, GM-CSF, IFN-Ɣ, IL-1β in cell supernatants although levels of TNF- α and IL-6 were slightly increased after BRP treatment. Levels of IL-4, IL-10 and TGF-β were also reduced by BRP. BRP significantly reduced the up-regulation promoted by LPS of transcription of genes in inflammatory signaling (Pdk1, Pak1, Nfkb1, Mtcp1, Gsk3b, Fos and Elk1) and of Il1β and Il1f9 (fold-change rate > 5), which were further confirmed by the inhibition of NF-κB and MAPK signaling pathways. Furthermore, the upstream adaptor MyD88 adaptor-like (Mal), also known as TIRAP, involved in TLR2 and TLR4 signaling, was down- regulated in BRP treated LPS-activated macrophages. Given that BRP inhibited multiple signaling pathways in macrophages involved in the inflammatory process activated by LPS, our data indicated that BRP is a noteworthy food-source for the discovery of new bioactive compounds and a potential candidate to attenuate exhacerbated inflammatory diseases.
Collapse
Affiliation(s)
- Bruno Bueno-Silva
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Dione Kawamoto
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Ellen S. Ando-Suguimoto
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Severino M. Alencar
- College of Agriculture “Luiz de Queiroz” (ESALQ/USP), Piracicaba, SP, Brazil
| | - Pedro L. Rosalen
- Piracicaba Dental School, University of Campinas–UNICAMP, Department of Physiologic Sciences, Piracicaba, SP, Brazil
| | - Marcia P. A. Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
- * E-mail:
| |
Collapse
|
24
|
Huang L, Sherchan P, Wang Y, Reis C, Applegate RL, Tang J, Zhang JH. Phosphoinositide 3-Kinase Gamma Contributes to Neuroinflammation in a Rat Model of Surgical Brain Injury. J Neurosci 2015; 35:10390-401. [PMID: 26203135 PMCID: PMC4510283 DOI: 10.1523/jneurosci.0546-15.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/26/2015] [Accepted: 06/02/2015] [Indexed: 12/31/2022] Open
Abstract
Neuroinflammation plays an important role in the pathophysiology of surgical brain injury (SBI). Phosphoinositide 3-kinase gamma (PI3Kγ), predominately expressed in immune and endothelial cells, activates multiple inflammatory responses. In the present study, we investigated the role of PI3Kγ and PI3Kγ-activated phosphodiesterase 3B (PDE3B) in neuroinflammation in a rat model of SBI. One hundred and fifty-two male Sprague Dawley rats (weight 280-350 g) were subjected to a partial right frontal lobe corticotomy model of SBI. A PI3Kγ pharmacological inhibitor (AS252424 or AS605240) was administered intraperitoneally. PI3Kγ siRNA, human recombinant active-PI3Kγ protein, or human recombinant active-PDE3B protein were administered intracerebroventricularly. Post-SBI assessments included neurobehavioral tests, brain water content, Western blot, and immunohistochemistry. Endogenous PI3Kγ levels were increased within peri-resection brain tissues after SBI, accompanied by increased brain water content and neurological functional deficits. There was a trend toward increased endogenous PDE3B phosphorylation after SBI. The selective PI3Kγ inhibitors AS252424 and AS605240 reduced brain water content surrounding corticotomy and improved neurological function after SBI. SBI increased and PI3Kγ inhibitor decreased levels of myeloperoxidase, cluster of differentiation 3, mast cell degranulation, E-selectin, and IL-1 in peri-resection brain tissues. Direct administration of human recombinant active-PI3Kγ protein and active-PDE3B protein countered the protective effect of AS252424. PI3Kγ siRNA reduced PI3Kγ levels, decreased brain water content within peri-resection brain tissues, and improved neurological function after SBI. Collectively, our findings suggest that PI3Kγ contributed to neuroinflammation after SBI. The use of selective PI3Kγ inhibitors may be a novel approach to ameliorating SBI via their anti-inflammation effects. Significance statement: Life-saving or elective neurosurgeries often involve unavoidable damages to neighboring, nondiseased brain tissues. Such surgical brain injury (SBI) is attributable exclusively to the neurosurgical procedure itself and may cause postoperative complications that exacerbate neurological function. Although the importance of this medical problem is fully acknowledged, intraoperative administration of adjunctive treatment such as steroids and mannitol to patients undergoing neurosurgery appear not to be efficient remedies for SBI. To date, the issue of perioperative neuroprotection specifically against SBI has not been well studied. Using a clinically relevant rat model of SBI, we are exploring a new neuroprotective strategy targeting phosphoinositide 3-kinase gamma (PI3Kγ). PI3Kγ activates multiple inflammatory responses. By attenuating neuroinflammation, selective PI3Kγ inhibition would limit postoperative complications and benefit neurological outcomes.
Collapse
Affiliation(s)
- Lei Huang
- Departments of Anesthesiology, Physiology and Pharmacology, and
| | | | - Yuechun Wang
- Physiology and Pharmacology, and Department of Physiology, School of Medicine, University of Jinan, Guangzhou 510632, China
| | | | | | | | - John H Zhang
- Departments of Anesthesiology, Physiology and Pharmacology, and Neurosurgery, Loma Linda University, Loma Linda, California 92354, and
| |
Collapse
|
25
|
Reddy NM, Potteti HR, Vegiraju S, Chen HJ, Tamatam CM, Reddy SP. PI3K-AKT Signaling via Nrf2 Protects against Hyperoxia-Induced Acute Lung Injury, but Promotes Inflammation Post-Injury Independent of Nrf2 in Mice. PLoS One 2015; 10:e0129676. [PMID: 26075390 PMCID: PMC4467869 DOI: 10.1371/journal.pone.0129676] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 05/12/2015] [Indexed: 12/16/2022] Open
Abstract
Lung epithelial and endothelial cell death accompanied by inflammation contributes to hyperoxia-induced acute lung injury (ALI). Impaired resolution of ALI can promote and/or perpetuate lung pathogenesis, including fibrosis. Previously, we have shown that the transcription factor Nrf2 induces cytoprotective gene expression and confers protection against hyperoxic lung injury, and that Nrf2-mediated signaling is also crucial for the restoration of lung homeostasis post-injury. Although we have reported that PI3K/AKT signaling is required for Nrf2 activation in lung epithelial cells, significance of the PI3K/AKT-Nrf2 crosstalk during hyperoxic lung injury and repair remains unclear. Thus, we evaluated this aspect using Nrf2 knockout (Nrf2–/–) and wild-type (Nrf2+/+) mouse models. Here, we show that pharmacologic inhibition of PI3K/AKT signaling increased lung inflammation and alveolar permeability in Nrf2+/+ mice, accompanied by decreased expression of Nrf2-target genes such as Nqo1 and Hmox1. PI3K/AKT inhibition dampened hyperoxia-stimulated Nqo1 and Hmox1 expression in lung epithelial cells and alveolar macrophages. Contrasting with its protective effects, PI3K/AKT inhibition suppressed lung inflammation in Nrf2+/+ mice during post-injury. In Nrf2–/– mice exposed to room-air, PI3K/AKT inhibition caused lung injury and inflammation, but it did not exaggerate hyperoxia-induced ALI. During post-injury, PI3K/AKT inhibition did not augment, but rather attenuated, lung inflammation in Nrf2–/– mice. These results suggest that PI3K/AKT-Nrf2 signaling is required to dampen hyperoxia-induced lung injury and inflammation. Paradoxically, the PI3K/AKT pathway promotes lung inflammation, independent of Nrf2, during post-injury.
Collapse
Affiliation(s)
- Narsa M. Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Haranatha R. Potteti
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Suryanarayana Vegiraju
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Hsin-Jou Chen
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Chandra Mohan Tamatam
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Sekhar P. Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
26
|
PI3K signalling in inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:882-97. [PMID: 25514767 DOI: 10.1016/j.bbalip.2014.12.006] [Citation(s) in RCA: 379] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/24/2014] [Accepted: 12/09/2014] [Indexed: 12/13/2022]
Abstract
PI3Ks regulate several key events in the inflammatory response to damage and infection. There are four Class I PI3K isoforms (PI3Kα,β,γ,δ), three Class II PI3K isoforms (PI3KC2α, C2β, C2γ) and a single Class III PI3K. The four Class I isoforms synthesise the phospholipid 'PIP3'. PIP3 is a 'second messenger' used by many different cell surface receptors to control cell movement, growth, survival and differentiation. These four isoforms have overlapping functions but each is adapted to receive efficient stimulation by particular receptor sub-types. PI3Kγ is highly expressed in leukocytes and plays a particularly important role in chemokine-mediated recruitment and activation of innate immune cells at sites of inflammation. PI3Kδ is also highly expressed in leukocytes and plays a key role in antigen receptor and cytokine-mediated B and T cell development, differentiation and function. Class III PI3K synthesises the phospholipid PI3P, which regulates endosome-lysosome trafficking and the induction of autophagy, pathways involved in pathogen killing, antigen processing and immune cell survival. Much less is known about the function of Class II PI3Ks, but emerging evidence indicates they can synthesise PI3P and PI34P2 and are involved in the regulation of endocytosis. The creation of genetically-modified mice with altered PI3K signalling, together with the development of isoform-selective, small-molecule PI3K inhibitors, has allowed the evaluation of the individual roles of Class I PI3K isoforms in several mouse models of chronic inflammation. Selective inhibition of PI3Kδ, γ or β has each been shown to reduce the severity of inflammation in one or more models of autoimmune disease, respiratory disease or allergic inflammation, with dual γ/δ or β/δ inhibition generally proving more effective. The inhibition of Class I PI3Ks may therefore offer a therapeutic opportunity to treat non-resolving inflammatory pathologies in humans. This article is part of a Special Issue entitled Phosphoinositides.
Collapse
|
27
|
Zhao K, Song X, Huang Y, Yao J, Zhou M, Li Z, You Q, Guo Q, Lu N. Wogonin inhibits LPS-induced tumor angiogenesis via suppressing PI3K/Akt/NF-κB signaling. Eur J Pharmacol 2014; 737:57-69. [DOI: 10.1016/j.ejphar.2014.05.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 04/28/2014] [Accepted: 05/07/2014] [Indexed: 10/25/2022]
|
28
|
Cho JS, Kang JH, Park IH, Lee HM. Steroids inhibit vascular endothelial growth factor expression via TLR4/Akt/NF-κB pathway in chronic rhinosinusitis with nasal polyp. Exp Biol Med (Maywood) 2014; 239:913-921. [DOI: 10.1177/1535370214537742] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is elevated in chronic rhinosinusitis with nasal polyps. Steroids have anti-inflammatory properties and are ideal candidates for treating chronic inflammatory airways. The aims of this study were to identify the inhibitory effects and mechanisms of steroids on lipopolysaccharide (LPS)-induced VEGF expression in nasal polyps. Nasal polyp-derived fibroblasts (NPDFs) were stimulated with LPS alone or with both LPS and steroids were used to determine the expression levels of toll-like receptor (TLR)-4, myeloid differentiation primary response gene 88 ( MyD88), and VEGF by using reverse transcription-polymerase chain reaction (RT-PCR). VEGF protein level was analyzed by immunocytochemical staining and enzyme-linked immunosorbent assay (ELISA). Small interfering RNA (siRNA) for TLR4 was transfected to down-regulate TLR4 expression. Activation of Akt and nuclear factor κB (NF-κB) pathway on VEGF expression was determined by Western blot analysis, immunocytochemical staining, and ELISA. Nasal polyp organ cultures were stimulated with LPS alone or in conjunction with steroids or LPS-RS (TLR4 inhibitor) and accessed the expression of VEGF. Steroids decreased the expressions of TLR4, MyD88, and VEGF mRNA and VEGF protein in LPS-stimulated NPDFs. Steroids inhibited LPS-induced VEGF expression levels in dose-dependent manner. The suppression of TLR4 transcription by siRNA treatment reduced LPS-induced expression of both TLR4 and VEGF in NPDFs. Furthermore, steroids inhibited the production of VEGF by blocking Akt and NF-κB activation and preventing with NF-κB translocation. Also, steroid and TLR4 inhibitor decreased VEGF expression in nasal polyp organ cultures. These results indicate that steroids inhibit LPS-induced VEGF expression through the TLR4/Akt/NF-κB signaling pathway in chronic rhinosinusitis with nasal polyp.
Collapse
Affiliation(s)
- Jung-Sun Cho
- Biomedical Science, Guro Hospital, Korea University College of Medicine, Seoul 152-703, Korea
- Institute for Medical Devices Clinical Trial Center, Guro Hospital, Korea University College of Medicine, Seoul 152-703, Korea
| | - Ju-Hyung Kang
- Biomedical Science, Guro Hospital, Korea University College of Medicine, Seoul 152-703, Korea
| | - Il-Ho Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul 152-703, Korea
| | - Heung-Man Lee
- Biomedical Science, Guro Hospital, Korea University College of Medicine, Seoul 152-703, Korea
- Institute for Medical Devices Clinical Trial Center, Guro Hospital, Korea University College of Medicine, Seoul 152-703, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul 152-703, Korea
| |
Collapse
|
29
|
Markou T, Chambers DJ. Lung injury after simulated cardiopulmonary bypass in an isolated perfused rat lung preparation: Role of mitogen-activated protein kinase/Akt signaling and the effects of theophylline. J Thorac Cardiovasc Surg 2014; 148:2335-44. [PMID: 24841445 PMCID: PMC4226635 DOI: 10.1016/j.jtcvs.2014.04.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/14/2014] [Accepted: 04/17/2014] [Indexed: 12/25/2022]
Abstract
Objectives Lung deflation and inflation during cardiac surgery with cardiopulmonary bypass contributes to pulmonary dysfunction postoperatively. Theophylline treatment for lung diseases has traditionally been thought to act by phosphodiesterase inhibition; however, increasing evidence has suggested other plausible mechanisms. We investigated the effects of deflation and reinflation on signaling pathways (p38-mitogen-activated protein kinase [MAPK], extracellular signal-regulated kinase 1 and 2 [ERK1/2], and Akt) and whether theophylline influences the deflation-induced lung injury and associated signaling. Methods Isolated rat lungs were perfused (15 mL/min) with deoxygenated rat blood in bicarbonate buffer and ventilated. After 20 minutes' equilibration, the lungs were deflated (60 minutes, aerobic perfusion 1.5 mL/min), followed by reinflation (60 minutes, anaerobic reperfusion 15 mL/min). Compliance, vascular resistance, and kinase phosphorylation were assessed during deflation and reinflation. The effects of SB203580 (50 μM), a p38-MAPK inhibitor, and theophylline (0.083 mM [therapeutic] or 3 mM [supratherapeutic]) on physiology and signaling were studied. Results Deflation reduced compliance by 44% compared with continuously ventilated lungs. p38-MAPK and Akt phosphorylation increased (three to fivefold) during deflation and reinflation, and ERK1/2 phosphorylation increased (approximately twofold) during reinflation. SB203580 had no effect on lung physiology or ERK1/2 and Akt activation. Both theophylline doses increased cyclic adenosine monophosphate, but only 3 mM theophylline improved compliance. p38-MAPK phosphorylation was not affected by theophylline; 0.083 mM theophylline inhibited reinflation-induced ERK1/2 phosphorylation (72% ± 3%); and 3 mM theophylline inhibited Akt phosphorylation during deflation (75% ± 5%) and reinflation (87% ± 4%). Conclusions Lung deflation and reinflation stimulates differential p38-MAPK, ERK1/2, and Akt activation, suggesting a role in lung injury during cardiopulmonary bypass. However, p38-MAPK was not involved in the compromised compliance. A supratherapeutic theophylline dose protected lungs against deflation-induced injury and was associated with inhibition of phosphoinositide 3-kinase/Akt rather than phosphodiesterase.
Collapse
Affiliation(s)
- Thomais Markou
- Cardiac Surgical Research, Rayne Institute, King's College London, St Thomas' Hospital Campus, London, United Kingdom.
| | - David J Chambers
- Cardiac Surgical Research, Rayne Institute, King's College London, Guy's and St Thomas' National Health Service Foundation Trust, St Thomas' Hospital, London, United Kingdom
| |
Collapse
|
30
|
Liang XM, Guo GF, Huang XH, Duan WL, Zeng ZL. Isotetrandrine protects against lipopolysaccharide-induced acute lung injury by suppression of mitogen-activated protein kinase and nuclear factor-kappa B. J Surg Res 2014; 187:596-604. [DOI: 10.1016/j.jss.2013.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/13/2013] [Accepted: 11/04/2013] [Indexed: 11/29/2022]
|
31
|
Xu F, Diao R, Liu J, Kang Y, Wang X, Shi L. Curcumin attenuatesstaphylococcus aureus-induced acute lung injury. CLINICAL RESPIRATORY JOURNAL 2014; 9:87-97. [PMID: 24460792 DOI: 10.1111/crj.12113] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 01/13/2014] [Accepted: 01/21/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Feng Xu
- Department of Infectious Diseases; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Ran Diao
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
- Center for Allergy; Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou China
| | - Jin Liu
- Department of Infectious Diseases; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Yanhua Kang
- Division of Immunology; Hangzhou Normal University; Hangzhou China
| | - Xuanding Wang
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Liyun Shi
- Division of Immunology; Hangzhou Normal University; Hangzhou China
| |
Collapse
|
32
|
Jin M, Zhou Q, Lee E, Dan S, Duan HQ, Kong D. AS252424, a PI3Kγ Inhibitor, Downregulates Inflammatory Responsiveness in Mouse Bone Marrow-Derived Mast Cells. Inflammation 2014; 37:1254-60. [DOI: 10.1007/s10753-014-9852-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
33
|
EGF-induced bronchial epithelial cells drive neutrophil chemotactic and anti-apoptotic activity in asthma. PLoS One 2013; 8:e72502. [PMID: 24039773 PMCID: PMC3770689 DOI: 10.1371/journal.pone.0072502] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/10/2013] [Indexed: 11/19/2022] Open
Abstract
Chronic damage and repair of the bronchial epithelium are features of asthma. We have previously reported that ex vivo stimulation of normal bronchial epithelial cells with epidermal growth factor (EGF), a key factor of epithelial repair, enhances the mechanisms of neutrophil accumulation, thereby promoting neutrophil defences during acute injury but potentially enhancing inflammation in chronic airway diseases. We have now sought to (i) determine whether this EGF-dependent pro-neutrophil activity is increased in asthma, where EGF and its epithelial receptor are over-expressed, and (ii) elucidate some of the mechanisms underlying this asthmatic epithelial-neutrophil interaction. Primary bronchial epithelial cells (PBEC) from healthy subjects, mild asthmatics and moderate-to-severe asthmatics (Mod/Sev) were stimulated with EGF, a model that mimics a repairing epithelium. Conditioned culture media (EGF-CM) were assessed for neutrophil chemotactic and anti-apoptotic activities and inflammatory mediator production. EGF induced the epithelium to produce soluble mediators with neutrophil chemotactic (p<0.001) and pro-survival (p = 0.021) activities which were related to the clinical severity of asthma (trend p = 0.010 and p = 0.009, respectively). This was associated with enhanced IL-6, IL-8, GM-CSF and TNF-α release, and cytokine-neutralising experiments using EGF-CM from Mod/Sev asthmatics demonstrated a role for GM-CSF in neutrophil survival (p<0.001). Pre-treatment of neutrophils with specific inhibitors of the myeloid-restricted class I phosphatidylinositol-3-OH kinase (PI(3)K) isoforms showed that the EGF-CM from Mod/Sev asthmatics depended on the γ (p<0.021) but not δ isoforms, while neutrophil survival required multiple class I PI(3)Ks. The EGF-induced chemotactic, but not pro-survival activity, involved RhoA signaling in neutrophils (p = 0.012). EGF whose activity is upregulated in asthma induces ex vivo the epithelium from asthmatic patients to produce pro-neutrophil activities; these are related to asthma severity and, in moderate-to-severe asthmatics, involves class IB PI(3)Kγ signaling, providing a potential therapeutic target for neutrophilic forms of asthma.
Collapse
|
34
|
Chen X, Yang X, Liu T, Guan M, Feng X, Dong W, Chu X, Liu J, Tian X, Ci X, Li H, Wei J, Deng Y, Deng X, Chi G, Sun Z. Kaempferol regulates MAPKs and NF-κB signaling pathways to attenuate LPS-induced acute lung injury in mice. Int Immunopharmacol 2012; 14:209-16. [DOI: 10.1016/j.intimp.2012.07.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/01/2012] [Accepted: 07/11/2012] [Indexed: 12/21/2022]
|