1
|
Xu Z, Chu M. Advances in Immunosuppressive Agents Based on Signal Pathway. Front Pharmacol 2022; 13:917162. [PMID: 35694243 PMCID: PMC9178660 DOI: 10.3389/fphar.2022.917162] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022] Open
Abstract
Immune abnormality involves in various diseases, such as infection, allergic diseases, autoimmune diseases, as well as transplantation. Several signal pathways have been demonstrated to play a central role in the immune response, including JAK/STAT, NF-κB, PI3K/AKT-mTOR, MAPK, and Keap1/Nrf2/ARE pathway, in which multiple targets have been used to develop immunosuppressive agents. In recent years, varieties of immunosuppressive agents have been approved for clinical use, such as the JAK inhibitor tofacitinib and the mTOR inhibitor everolimus, which have shown good therapeutic effects. Additionally, many immunosuppressive agents are still in clinical trials or preclinical studies. In this review, we classified the immunosuppressive agents according to the immunopharmacological mechanisms, and summarized the phase of immunosuppressive agents.
Collapse
Affiliation(s)
- Zhiqing Xu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Pharmacology, Jilin University, Changchun, China
| | - Ming Chu
- Department of Immunology, National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
2
|
Luo T, Jiang S, Zhou B, Song Q, Du J, Liu P, Wang X, Song H, Shao C. Protective Effect of Isoorientin on Oleic Acid-Induced Oxidative Damage and Steatosis in Rat Liver Cells. Front Pharmacol 2022; 13:818159. [PMID: 35185572 PMCID: PMC8853441 DOI: 10.3389/fphar.2022.818159] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/03/2022] [Indexed: 12/18/2022] Open
Abstract
The harm of nonalcoholic fatty liver disease to human health is increasing, which calls for urgent prevention and treatment of the disease. Isoorientin is an effective ingredient of Chinese herbal medicine with anti-inflammatory and antioxidant effects. However, the effect of isoorientin in nonalcoholic fatty liver disease is still unclear. In this study, combined in vivo and in vitro experiments, through pathological observation, flow cytometry, immunofluorescence and western blot analysis to explore the role of isoorientin in steatosis and reveal its molecular mechanism. The results demonstrated that oleic acid treatment significantly increased the content of ROS and lipid droplets in rat hepatocytes, and promoted the expression of γH2AX, HO-1, PPARγ, SREBP-1c, FAS. The ROS content in the cells of co-treated with isoorientin and oleic acid was significantly reduced compared to the oleic acid group, and the expression of γH2AX, HO-1, PPARγ, SREBP-1c, FAS, and the nuclear translocation of NF-κB p65 were also significantly inhibited. Our data showed that oleic acid induce oxidative damage and steatosis in hepatocytes both in vitro and in vivo, and activate the PPARγ/NF-κB p65 signal pathway. Moreover, isoorientin can significantly reduce oleic acid -induced oxidative damage and steatosis by regulating the PPARγ/NF-kB p65 signal pathway.
Collapse
Affiliation(s)
- Tongwang Luo
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
- *Correspondence: Tongwang Luo, ; Houhui Song, ; Chunyan Shao,
| | - Sheng Jiang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Bin Zhou
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Quanjiang Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Jing Du
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Ping Liu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Xiaodu Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Houhui Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
- *Correspondence: Tongwang Luo, ; Houhui Song, ; Chunyan Shao,
| | - Chunyan Shao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, Hangzhou, China
- Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Hangzhou, China
- China-Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, China
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
- *Correspondence: Tongwang Luo, ; Houhui Song, ; Chunyan Shao,
| |
Collapse
|
3
|
Hong W, Zhi FX, Kun TH, Hua FJ, Huan Ling L, Fang F, Wen C, Jie W, Yang LC. 6-Gingerol attenuates ventilator-induced lung injury via anti-inflammation and antioxidative stress by modulating the PPARγ/NF-κBsignalling pathway in rats. Int Immunopharmacol 2021; 92:107367. [PMID: 33461160 DOI: 10.1016/j.intimp.2021.107367] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023]
Abstract
Although mechanical ventilation (MV) is indispensable to life-support therapy in critically ill patients, it may promote or aggravatelunginjury known asventilator-inducedlunginjury(VILI). 6-Gingerol is the principal ingredient of ginger with potential anti-inflammatory and antioxidant properties in various diseases. Nevertheless, the role and mechanism of 6-gingerol in the process of VILI has not been explicitly investigated. In the study, we found that pre-treatment with 6-gingerol significantly improved the histological changes and pulmonary oedema, inhibited neutrophil accumulation and the release of early pro-inflammatory cytokines and MPO, and reduced oxidative stress reactions after high MV. Moreover, 6-gingerol treatment also increased PPARγ expression and decreased NF-κB activation in rats subjected to high MV. Furthermore, GW9662, a specific PPARγ inhibitor, was demonstrated to activatethe NF-κB pathway and cancele the protective role of 6-gingerol in VILI. This indicates that 6-gingerol exerted anti-inflammatory and antioxidative stress effects in VILI by activating PPARγ and inhibiting the NF-κBsignalling pathway.
Collapse
Affiliation(s)
- Wei Hong
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Fang Xiang Zhi
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Anesthesiology, Subei People's Hospital, YangZhou 225001, China
| | - Tu Han Kun
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Feng Jie Hua
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Li Huan Ling
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Fang Fang
- Department of General Medicine, Huazhong University of Science and Technology Union ShenZhen Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Chen Wen
- Department of Anesthesiology, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, Guangdong Province, China
| | - Wang Jie
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Li Chao Yang
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China.
| |
Collapse
|
4
|
Meng C, Qian Y, Zhang C, Liu H, Mu X, Zhang A. IKKε deficiency inhibits acute lung injury following renal ischemia reperfusion injury. Mol Med Rep 2020; 22:4213-4220. [PMID: 33000218 PMCID: PMC7533469 DOI: 10.3892/mmr.2020.11532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
Renal ischemia reperfusion injury (IRI) after surgery may promote acute lung injury (ALI) by inducing an inflammatory response. However, the underlying molecular mechanism is still unclear. Studies have reported that inhibitor of κB kinase (IKK)ε primarily regulates inflammation and cell proliferation. The present study aimed to investigate the regulatory role of IKKε in ALI in mice, in order to provide an experimental basis for preventing ALI following surgery-induced renal IRI. C57BL/6J wild-type (WT) and IKKε knockout (IKKε−/−) mice underwent bilateral renal pedicle occlusion. The plasma creatinine concentration, urea nitrogen level and lung wet-to-dry ratio were measured at baseline, and at 24 and 48 h after declamping. The histological localization and protein levels of inflammatory factors, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10, were analyzed in lung tissues. Subsequently, the interactions between IKKε and components of the nuclear factor (NF)-κB pathway were studied. The results of the present study demonstrated that the IKKε−/− groups displayed similar renal function but less pulmonary edema compared with that of the WT groups. The levels of proinflammatory factors in the lungs were significantly upregulated in WT mice compared with those in IKKε−/− mice after IRI surgery. The NF-κB pathway components and downstream factors were substantially upregulated in the WT groups after acute ischemic kidney injury, and these effects were significantly inhibited in the IKKε−/− groups. Based on these data, the present study hypothesized that IKKε may serve a negative role in kidney-lung crosstalk after renal IRI and may be a novel target for the treatment of patients with renal IRI.
Collapse
Affiliation(s)
- Chao Meng
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| | - Yi Qian
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Cui Zhang
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| | - Han Liu
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| | - Xinwei Mu
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| | - Aiping Zhang
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210001, P.R. China
| |
Collapse
|
5
|
Fang XZ, Ge YL, Chen ZY, Shu HQ, Yang YY, Yu Y, Zhou XJ, Chen L, Cui SN, Wang YX, Yao SL, Shang Y. NecroX-5 alleviate lipopolysaccharide-induced acute respiratory distress syndrome by inhibiting TXNIP/NLRP3 and NF-κB. Int Immunopharmacol 2020; 81:106257. [PMID: 32044659 DOI: 10.1016/j.intimp.2020.106257] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/04/2020] [Accepted: 01/24/2020] [Indexed: 02/07/2023]
Abstract
The activation of NLRP3 inflammasome and NF-κB pathway, associating with oxidativestress, have been implicated in the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). NecroX-5 has been reported to exhibit theeffectsofanti-oxidation and anti-stress in various diseases. However, the role of NecroX-5 in ALI has not been explicitly demonstrated. The aim of this study was to explore the therapeutic effects and potential mechanism action of NecroX-5 on ALI. Here, we found that NecroX-5 pretreatment dramatically diminished the levels of IL-1β, IL-18 and ROS in in RAW264.7 cells challenged with LPS and ATP. Furthermore, NecroX-5 suppressed the activation of NLRP3 inflammasome and NF-κB signalpathway. In addition, NecroX-5 also inhibited the thioredoxin-interacting protein (TXNIP) expression. In vivo, NecroX-5 reduced the LPS-induced lung histopathological injury, the number of TUNEL-positive cells, lung wet/dry (W/D) ratio, levels of total protein and inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) in mice. Additionally, LPS-induced upregulation of myeloperoxidase (MPO), ROS production and malondialdehyde (MDA) were inhibited by NecroX-5 administration. Thus, our results demonstrate that NecroX-5 protects against LPS-induced ALI by inhibiting TXNIP/NLRP3 and NF-κB.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Department of Anesthesiology, Clinical Medical School of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, Jiangsu Province, PR China
| | - Ya-Li Ge
- Department of Anesthesiology, Clinical Medical School of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, Jiangsu Province, PR China
| | - Zhao-Yuan Chen
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hua-Qing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yi-Yi Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong Universityof Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xiao-Jing Zhou
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lin Chen
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shu-Nan Cui
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shang-Long Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong Universityof Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, Hubei 430022, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College,Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| |
Collapse
|
6
|
Novel Synthetic, Host-defense Peptide Protects Against Organ Injury/Dysfunction in a Rat Model of Severe Hemorrhagic Shock. Ann Surg 2019; 268:348-356. [PMID: 28288070 DOI: 10.1097/sla.0000000000002186] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To evaluate (1) levels of the host-defense/antimicrobial peptide LL-37 in patients with trauma and hemorrhagic shock (HS) and (2) the effects of a synthetic host-defense peptide; Pep19-4LF on multiple organ failure (MOF) associated with HS. BACKGROUND HS is a common cause of death in severely injured patients. There is no specific therapy that reduces HS-associated MOF. METHODS (1) LL-37 was measured in 47 trauma/HS patients admitted to an urban major trauma center. (2) Male Wistar rats were submitted to HS (90 min, target mean arterial pressure: 27-32 mm Hg) or sham operation. Rats were treated with Pep19-4LF [66 (n = 8) or 333 μg/kg · h (n = 8)] or vehicle (n = 12) for 4 hours following resuscitation. RESULTS Plasma LL-37 was 12-fold higher in patients with trauma/HS compared to healthy volunteers. HS rats treated with Pep19-4LF (high dose) had a higher mean arterial pressure at the end of the 4-hour resuscitation period (79 ± 4 vs 54 ± 5 mm Hg) and less renal dysfunction, liver injury, and lung inflammation than HS rats treated with vehicle. Pep19-4LF enhanced (kidney/liver) the phosphorylation of (1) protein kinase B and (2) endothelial nitric oxide synthase. Pep19-4LF attenuated the HS-induced (1) translocation of p65 from cytosol to nucleus, (2) phosphorylation of IκB kinase on Ser, and (3) phosphorylation of IκBα on Ser resulting in inhibition of nuclear factor kappa B and formation of proinflammatory cytokines. Pep19-4LF prevented the release of tumor necrosis factor alpha caused by heparan sulfate in human mononuclear cells by binding to this damage-associated molecular pattern. CONCLUSIONS Trauma-associated HS results in release of LL-37. The synthetic host-defense/antimicrobial peptide Pep19-4LF attenuates the organ injury/dysfunction associated with HS.
Collapse
|
7
|
Li L, Cataisson C, Flowers B, Fraser E, Sanchez V, Day CP, Yuspa SH. Topical Application of a Dual ABC Transporter Substrate and NF-κB Inhibitor Blocks Multiple Sources of Cutaneous Inflammation in Mouse Skin. J Invest Dermatol 2019; 139:1506-1515.e7. [PMID: 30684549 DOI: 10.1016/j.jid.2018.12.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/21/2018] [Accepted: 12/30/2018] [Indexed: 12/16/2022]
Abstract
Among the molecular signals underlying cutaneous inflammation is the transcription complex NF-κB, its upstream modulators, and cytokines and chemokines that are the downstream proinflammatory effectors. Central to NF-κB activation is IκB kinase (IKK), which phosphorylates IκBα, releasing NF-κB to the nucleus. In a screening of a kinase inhibitor library, we identified two IKK inhibitors that were high-affinity substrates for p-glycoprotein (ABCB1), the multidrug resistance protein known to facilitate transdermal drug delivery. ACHP (2-amino-6-[2-(cyclopropylmethoxy)-6-hydroxyphenyl]-4-(4-piperidinyl)-3-pyridinecarbonitrile) and IKK 16 prevented both nuclear translocation of NF-κB and activation of a NF-κB reporter and reduced the induction of cytokine and chemokine transcripts in human or mouse keratinocytes by IL-1α, tumor necrosis factor-α, and phorbol myristate acetate. ACHP, but not IKK 16, was nontoxic to mouse or human keratinocytes at any dose tested. In mice, topical ACHP prevented the cutaneous inflammation induced by topical phorbol myristate acetate or imiquimod, reduced the inflammation from erythema doses of artificial sunlight, and lowered the tumor incidence of mice treated with 7,12-dimethyl benzanthracene when applied before phorbol myristate acetate. Topical ACHP also reduced the NF-κB and IL-17 inflammatory signature after multiple doses of imiquimod. Thus, ACHP and IKK 16 hit their NF-κB target in mouse and human keratinocytes, and ACHP is an effective topical nonsteroidal anti-inflammatory in mice.
Collapse
Affiliation(s)
- Luowei Li
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Brittany Flowers
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Elise Fraser
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Vanesa Sanchez
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
| |
Collapse
|
8
|
Wan B, Xu WJ, Zhan P, Jin JJ, Xi GM, Chen MZ, Hu YB, Zhu SH, Liu HB, Wang XX, Zhang XW, Lv TF, Song Y. Topotecan alleviates ventilator-induced lung injury via NF-κB pathway inhibition. Cytokine 2018; 110:381-388. [DOI: 10.1016/j.cyto.2018.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/26/2018] [Accepted: 04/15/2018] [Indexed: 11/25/2022]
|
9
|
Prescott JA, Cook SJ. Targeting IKKβ in Cancer: Challenges and Opportunities for the Therapeutic Utilisation of IKKβ Inhibitors. Cells 2018; 7:cells7090115. [PMID: 30142927 PMCID: PMC6162708 DOI: 10.3390/cells7090115] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/15/2018] [Accepted: 08/19/2018] [Indexed: 02/08/2023] Open
Abstract
Deregulated NF-κB signalling is implicated in the pathogenesis of numerous human inflammatory disorders and malignancies. Consequently, the NF-κB pathway has attracted attention as an attractive therapeutic target for drug discovery. As the primary, druggable mediator of canonical NF-κB signalling the IKKβ protein kinase has been the historical focus of drug development pipelines. Thousands of compounds with activity against IKKβ have been characterised, with many demonstrating promising efficacy in pre-clinical models of cancer and inflammatory disease. However, severe on-target toxicities and other safety concerns associated with systemic IKKβ inhibition have thus far prevented the clinical approval of any IKKβ inhibitors. This review will discuss the potential reasons for the lack of clinical success of IKKβ inhibitors to date, the challenges associated with their therapeutic use, realistic opportunities for their future utilisation, and the alternative strategies to inhibit NF-κB signalling that may overcome some of the limitations associated with IKKβ inhibition.
Collapse
Affiliation(s)
- Jack A Prescott
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
10
|
SN50, a Cell-Permeable Inhibitor of Nuclear Factor-κB, Attenuates Ventilator-Induced Lung Injury in an Isolated and Perfused Rat Lung Model. Shock 2018; 46:194-201. [PMID: 26780513 DOI: 10.1097/shk.0000000000000563] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High tidal volume (VT) ventilation causes the release of various mediators and results in ventilator-induced lung injury (VILI). SN50, a cell-permeable nuclear factor-κB (NF-κB) inhibitory peptide, attenuates inflammation and acute respiratory distress syndrome. However, the mechanisms associated with the effects of SN50 in VILI have not been fully elucidated. We investigated the cellular and molecular mechanisms for the effects of SN50 treatment in VILI. An isolated and perfused rat lung model was exposed to low (5 mL/kg) or high (15 mL/kg) VT ventilation for 6 h. SN50 was administered in the perfusate at the onset of the high-stretch mechanical ventilation. The hemodynamics, lung histological changes, inflammatory responses, and activation of apoptotic pathways were evaluated. VILI was demonstrated by increased pulmonary vascular permeability and lung weight gain, as well as by increased levels of interleukin (IL)-1β, tumor necrosis factor (TNF)-α, myeloperoxidase (MPO), hydrogen peroxide, and macrophage inflammatory protein-2 in the bronchoalveolar lavage fluid. The lung tissue expression of TNF-α, IL-1β, mitogen-activated protein kinases (MAPKs), caspase-3, and phosphorylation of serine/threonine-specific protein kinase (p-AKT) was greater in the high VT group than in the low VT group. Upregulation and activation of NF-κB was associated with increased lung injury in VILI. SN50 attenuated the inflammatory responses, including the expression of IL-1β, TNF-α, MPO, MAPKs, and NF-κB. In addition, the downregulation of apoptosis was evaluated using caspase-3 and p-AKT expression. Furthermore, SN50 mitigated the increases in the lung weights, pulmonary vascular permeability, and lung injury. In conclusion, VILI is associated with inflammatory responses and activation of NF-κB. SN50 inhibits the activation of NF-κB and attenuates VILI.
Collapse
|
11
|
Fang XZ, Ge YL, Li M, Huang TF, Yang Z, Gao J. Preconditioning of physiological cyclic stretch inhibits the inflammatory response induced by pathologically mechanical stretch in alveolar epithelial cells. Exp Ther Med 2017; 15:2172-2176. [PMID: 29434821 DOI: 10.3892/etm.2017.5611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 04/28/2017] [Indexed: 01/09/2023] Open
Abstract
The aim of the present study was to investigate the effects of preconditioning of physiological cyclic stretch (CS) on the overexpression of early pro-inflammatory cytokines [including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-8] during the inflammatory response induced by pathologically mechanical stretch in lung epithelial cells, and to determine its molecular mechanism of action. Cells were subjected to 5% CS for various durations (0, 15, 30, 60 and 120 min) prior to 6 h treatment with pathological 20% CS. In a separate experiment, cells were preconditioned with physiological 5% CS or incubated with a nuclear factor (NF)-κB inhibitor, pyrroldine dithiocarbamate (PDTC). The expression levels of inflammatory mediators were measured using reverse transcription-quantitative polymerase chain reaction. NF-κB was quantified using western blot analysis. Preconditioning with physiological 5% CS for 30, 60 and 120 min was demonstrated to significantly attenuate the release of pathologically mechanical stretch-induced early pro-inflammatory cytokines (TNF-α, IL-1β and IL-8) in alveolar epithelial cells (P<0.05) and significantly reduce the expression of NF-κB (P<0.05). Peak suppression was observed in cells preconditioned for 60 min. In the second set of experiments, it was demonstrated that mechanical stretch-induced release of TNF-α, IL-1β and IL-8 was significantly inhibited by both PDTC pretreatment and 5% CS pretreatment alone (all P<0.05). Furthermore, significant inhibition was also observed when both 5% CS pretreatment and PDTC pretreatment was used on mechanical stretch-induced cells (P<0.05), which was markedly greater than the inhibition induced by either pretreatment alone. The present findings suggest that preconditioning with physiological 5% CS is able to inhibit the inflammatory response induced by pathologically mechanical stretch in alveolar epithelial cells. These anti-inflammatory effects are induced, at least in part, by suppressing the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Ya-Li Ge
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Min Li
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Jiangsu 212000, P.R. China
| | - Tian-Feng Huang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Zhang Yang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Ju Gao
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
12
|
IκK-16 decreases miRNA-155 expression and attenuates the human monocyte inflammatory response. PLoS One 2017; 12:e0183987. [PMID: 28910312 PMCID: PMC5598939 DOI: 10.1371/journal.pone.0183987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/15/2017] [Indexed: 12/31/2022] Open
Abstract
Excessive inflammatory responses in the surgical patient may result in cellular hypo-responsiveness, which is associated with an increased risk of secondary infection and death. microRNAs (miRNAs), such as miR-155, are powerful regulators of inflammatory signalling pathways including nuclear factor κB (NFκB). Our objective was to determine the effect of IκK-16, a selective blocker of inhibitor of kappa-B kinase (IκK), on miRNA expression and the monocyte inflammatory response. In a model of endotoxin tolerance using primary human monocytes, impaired monocytes had decreased p65 expression with suppressed TNF-α and IL-10 production (P < 0.05). miR-155 and miR-138 levels were significantly upregulated at 17 h in the impaired monocyte (P < 0.05). Notably, IκK-16 decreased miR-155 expression with a corresponding dose-dependent decrease in TNF-α and IL-10 production (P < 0.05), and impaired monocyte function was associated with increased miR-155 and miR-138 expression. In the context of IκK-16 inhibition, miR-155 mimics increased TNF-α production, while miR-155 antagomirs decreased both TNF-α and IL-10 production. These data demonstrate that IκK-16 treatment attenuates the monocyte inflammatory response, which may occur through a miR-155-mediated mechanism, and that IκK-16 is a promising approach to limit the magnitude of an excessive innate inflammatory response to LPS.
Collapse
|
13
|
Li Q, Ge YL, Li M, Fang XZ, Yuan YP, Liang L, Huang SQ. miR-127 contributes to ventilator-induced lung injury. Mol Med Rep 2017; 16:4119-4126. [PMID: 28765901 DOI: 10.3892/mmr.2017.7109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 04/27/2017] [Indexed: 11/06/2022] Open
Abstract
Although it is essential in critical care medicine, mechanical ventilation often results in ventilator‑induced lung injury (VILI). Treating mice with lipopolysaccharide has been reported to upregulate the expression of miR‑127, which has been implicated in the modulation of immune responses. However, the putative roles of miR‑127 during the development of VILI have yet to be elucidated. The present study demonstrated that challenging mice with mechanical ventilation for 6 h significantly upregulated the expression of miR‑127 in bronchoalveolar lavage fluid, serum and lung tissue samples. Conversely, following the downregulation of miR‑127 expression in vivo using an adenovirus delivery system, VILI‑associated pathologies, including alterations in the pulmonary wet/dry ratio, pulmonary permeability, lung neutrophil infiltration and levels of pro‑inflammatory cytokines, were significantly attenuated. In addition, miR‑127 knockdown inhibited the ventilation‑induced activation of nuclear factor (NF)‑κB and p38 mitogen‑activated protein kinase (MAPK). These findings suggested that the upregulation of miR‑127 expression may contribute to the development of VILI, through the modulation of pulmonary permeability, the induction of histopathological alterations, and the potentiation of inflammatory responses involving NF‑κB and p38 MAPK‑associated signaling pathways.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| | - Ya-Li Ge
- Department of Anesthesiology, Subei People's Hospital of Jiangsu, Yangzhou, Jiangsu 225001, P.R. China
| | - Min Li
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Xiang-Zhi Fang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu, Yangzhou, Jiangsu 225001, P.R. China
| | - Yan-Ping Yuan
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| | - Lei Liang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Shao-Qiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| |
Collapse
|
14
|
Sordi R, Nandra KK, Chiazza F, Johnson FL, Cabrera CP, Torrance HD, Yamada N, Patel NSA, Barnes MR, Brohi K, Collino M, Thiemermann C. Artesunate Protects Against the Organ Injury and Dysfunction Induced by Severe Hemorrhage and Resuscitation. Ann Surg 2017; 265:408-417. [PMID: 28059970 DOI: 10.1097/sla.0000000000001664] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To evaluate the effects of artesunate on organ injury and dysfunction associated with hemorrhagic shock (HS) in the rat. BACKGROUND HS is still a common cause of death in severely injured patients and is characterized by impairment of organ perfusion, systemic inflammatory response, and multiple organ failure. There is no specific therapy that reduces organ injury/dysfunction. Artesunate exhibits pharmacological actions beyond its antimalarial activity, such as anticancer, antiviral, and anti-inflammatory effects. METHODS Rats were submitted to HS. Mean arterial pressure was reduced to 30 mm Hg for 90 minutes, followed by resuscitation. Rats were randomly treated with artesunate (2.4 or 4.8 mg/kg i.v.) or vehicle upon resuscitation. Four hours later, parameters of organ injury and dysfunction were assessed. RESULTS Artesunate attenuated the multiple organ injury and dysfunction caused by HS. Pathway analysis of RNA sequencing provided good evidence to support an effect of artesunate on the Akt-survival pathway, leading to downregulation of interleukin-1 receptor-associated kinase 1. Using Western blot analysis, we confirmed that treatment of HS rats with artesunate enhanced the phosphorylation (activation) of Protein kinase B (Akt) and endothelial nitric oxide synthase and the phosphorylation (inhibition) of glycogen synthase kinase-3β (GSK-3β). Moreover, artesunate attenuated the HS-induced activation of nuclear factor kappa B and reduced the expression of proinflammatory proteins (inducible nitric oxide synthase, tumor necrosis factor-α, and interleukin 6). CONCLUSIONS Artesunate attenuated the organ injury/dysfunction associated with HS by a mechanism that involves the activation of the Akt-endothelial nitric oxide synthase survival pathway, and the inhibition of glycogen synthase kinase-3β and nuclear factor kappa B. A phase II clinical trial evaluating the effects of good manufacturing practice-artesunate in patients with trauma and severe hemorrhage is planned.
Collapse
Affiliation(s)
- Regina Sordi
- *Centre for Translational Medicine and Therapeutics, Queen Mary University of London, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, London, UK †Department of Drug Science and Technology, University of Turin, Turin, Italy ‡Department of Clinical Pharmacology, Queen Mary University of London, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, London, UK §Centre for Trauma Sciences, Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine & Dentistry, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Chen X, Zhang X, Zhang J, Gao Y, Yang Z, Li S, Dai H. Attenuation of acute lung injury in a rat model by Semen Cassiae. Altern Ther Health Med 2017; 17:234. [PMID: 28454544 PMCID: PMC5408400 DOI: 10.1186/s12906-017-1747-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/20/2017] [Indexed: 12/29/2022]
Abstract
Background Acute lung injury (ALI) is an inflammatory disorder. Semen Cassiae has potent anti-inflammatory activities. The aim of our study was to investigate whether Semen Cassiae plays a protective effect on lipopolysaccharide (LPS)-induced ALI and, if so, to elucidate its potential mechanism. Methods Male Sprague–Dawley rat lungs were injured by intratracheal instillation of LPS. Rats were treated with Semen Cassiae or vehicle 3 h after LPS challenge. Samples were harvested 24 h post-LPS administration. We also investigated the effects of Semen Cassiae on LPS stimulation in RAW 264.7 cells. Results LPS administration markedly induced pulmonary edema and polymorphonuclear neutrophil influxes. These changes were significantly attenuated in Semen Cassiae treated group. Moreover, Semen Cassiae markedly reduced pulmonary interleukin (IL)-6, tumor necrosis factor (TNF)-α, and 8-hydroxy-2′-deoxyguanosine (8-OHdG) levels. The pulmonary soluble epoxide hydrolase (sEH) activity and the DNA binding activity of Nuclear factor (NF)-κB were significantly inhibited in Semen Cassiae treated group. Furthermore, Semen Cassiae treatment significantly increased epoxyeicosatrienoic acids (EETs), and heme oxygenase-1 (HO-1) activity. Our in vitro study demonstrates that Semen Cassiae treatment may inhibit LPS induced IκBα phosphorylation and NF-κB p65 nucleus translocation. Conclusion Semen Cassiae protects LPS-induced ALI in rats. Semen Cassiae can be developed as a novel treatment for ALI. Electronic supplementary material The online version of this article (doi:10.1186/s12906-017-1747-7) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Chen J, Kieswich JE, Chiazza F, Moyes AJ, Gobbetti T, Purvis GSD, Salvatori DCF, Patel NSA, Perretti M, Hobbs AJ, Collino M, Yaqoob MM, Thiemermann C. IκB Kinase Inhibitor Attenuates Sepsis-Induced Cardiac Dysfunction in CKD. J Am Soc Nephrol 2017; 28:94-105. [PMID: 27153924 PMCID: PMC5198262 DOI: 10.1681/asn.2015060670] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 03/09/2016] [Indexed: 01/01/2023] Open
Abstract
Patients with CKD requiring dialysis have a higher risk of sepsis and a 100-fold higher mortality rate than the general population with sepsis. The severity of cardiac dysfunction predicts mortality in patients with sepsis. Here, we investigated the effect of preexisting CKD on cardiac function in mice with sepsis and whether inhibition of IκB kinase (IKK) reduces the cardiac dysfunction in CKD sepsis. Male C57BL/6 mice underwent 5/6 nephrectomy, and 8 weeks later, they were subjected to LPS (2 mg/kg) or sepsis by cecal ligation and puncture (CLP). Compared with sham operation, nephrectomy resulted in significant increases in urea and creatinine levels, a small (P<0.05) reduction in ejection fraction (echocardiography), and increases in the cardiac levels of phosphorylated IκBα, Akt, and extracellular signal-regulated kinase 1/2; nuclear translocation of the NF-κB subunit p65; and inducible nitric oxide synthase (iNOS) expression. When subjected to LPS or CLP, compared with sham-operated controls, CKD mice exhibited exacerbation of cardiac dysfunction and lung inflammation, greater increases in levels of plasma cytokines (TNF-α, IL-1β, IL-6, and IL-10), and greater increases in the cardiac levels of phosphorylated IKKα/β and IκBα, nuclear translocation of p65, and iNOS expression. Treatment of CKD mice with an IKK inhibitor (IKK 16; 1 mg/kg) 1 hour after CLP or LPS administration attenuated these effects. Thus, preexisting CKD aggravates the cardiac dysfunction caused by sepsis or endotoxemia in mice; this effect may be caused by increased cardiac NF-κB activation and iNOS expression.
Collapse
Affiliation(s)
- Jianmin Chen
- Center for Translational Medicine and Therapeutics and
| | | | - Fausto Chiazza
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Amie J Moyes
- Center for Translational Medicine and Therapeutics and
| | - Thomas Gobbetti
- Center for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Daniela C F Salvatori
- Central Laboratory Animal Facility, Leiden University Medical Center, Leiden, The Netherlands; and
| | | | - Mauro Perretti
- Center for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Muhammad M Yaqoob
- Center for Translational Medicine and Therapeutics and
- Department of Renal Medicine and Transplantation, Royal London Hospital, Whitechapel, London, United Kingdom
| | | |
Collapse
|
17
|
Effects of a Soluble Epoxide Hydrolase Inhibitor on Lipopolysaccharide-Induced Acute Lung Injury in Mice. PLoS One 2016; 11:e0160359. [PMID: 27490848 PMCID: PMC4973880 DOI: 10.1371/journal.pone.0160359] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/18/2016] [Indexed: 01/02/2023] Open
Abstract
Objectives Inflammation plays a key role in the pathogenesis of acute lung injury (ALI). Soluble epoxide hydrolase (sEH) is suggested as a vital pharmacologic target for inflammation. In this study, we determined whether a sEH inhibitor, AUDA, exerts lung protection in lipopolysaccharide (LPS)-induced ALI in mice. Methods Male BALB/c mice were randomized to receive AUDA or vehicle intraperitoneal injection 4 h after LPS or phosphate buffered saline (PBS) intratracheal instillation. Samples were harvested 24 h post LPS or PBS administration. Results AUDA administration decreased the pulmonary levels of monocyte chemoattractant protein (MCP)-1 and tumor necrosis factor (TNF)-α. Improvement of oxygenation and lung edema were observed in AUDA treated group. AUDA significantly inhibited sEH activity, and elevated epoxyeicosatrienoic acids (EETs) levels in lung tissues. Moreover, LPS induced the activation of nuclear factor (NF)-κB was markedly dampened in AUDA treated group. Conclusion Administration of AUDA after the onset of LPS-induced ALI increased pulmonary levels of EETs, and ameliorated lung injury. sEH is a potential pharmacologic target for ALI.
Collapse
|
18
|
Huang J, Wu B, Song J, Wu J. Mechanisms of hyperventilation-induced lung injuries in neonatal rats. Minerva Pediatr (Torino) 2016; 73:73-79. [PMID: 27270839 DOI: 10.23736/s2724-5276.16.04458-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The aim of this study was to investigate the mechanism of early inflammatory injury in neonatal ventilator-induced lung injuries (VILI). METHODS Newborn rats were randomly assigned to groups and administrated mechanical ventilation with different tidal volumes. Morphological changes in lung tissues were observed, and the levels of interleukin-6 (IL-6), cysteinyl leukotriene mRNA (CysLT1 mRNA), and nuclear factor-κB mRNA (NF-κBp65 mRNA) in lung tissues were analyzed. RESULTS The ventilation groups exhibited different degrees of inflammatory cell infiltration, which was aggravated as the tidal volume and ventilation time increased. The IL-6 levels of the hyperventilation 5H, conventional ventilation 5H, hyperventilation 3H, control, and normal lung-tissue group were 785.33±39.06, 701.6±33.65, 686.65±46.85, 637.63±40.55, and 635.02±65.78 pg/g, respectively. Hyperventilation increased the levels of IL-6 and NF-κBp65 mRNA as the ventilation time increased, and IL-6 was positively correlated with NF-κBp65 mRNA levels (r=0.72, P<0.01). Longer hyperventilation periods upregulate the level of CysLT1 mRNA. CysLT1 mRNA/GAPDH of the hyperventilation 5H group was 2.14±1.45 (P<0.01). CONCLUSIONS Mechanical ventilation with a large tidal volume can cause VILI, characterized at an early stage by inflammatory responses and particularly by the increased secretion and invasion of inflammatory cytokines and inflammatory cells. The activation of the NF-κB-IL-6 signaling pathway was an important mechanism for the initiation of VILI. Additionally, CysLT1 was involved in the inflammatory VILI damage, and its upregulation occurred later than that of IL-6.
Collapse
Affiliation(s)
- Jinjie Huang
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Benqing Wu
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China -
| | - Jinzhi Song
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Jun Wu
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
19
|
Shu Y, Liu Y, Li X, Cao L, Yuan X, Li W, Cao Q. Aspirin-Triggered Resolvin D1 Inhibits TGF-β1-Induced EndMT through Increasing the Expression of Smad7 and Is Closely Related to Oxidative Stress. Biomol Ther (Seoul) 2016; 24:132-9. [PMID: 26869523 PMCID: PMC4774493 DOI: 10.4062/biomolther.2015.088] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/04/2015] [Accepted: 11/12/2015] [Indexed: 01/10/2023] Open
Abstract
The endothelial-mesenchymal transition (EndMT) is known to be involved in the transformation of vascular endothelial cells to mesenchymal cells. EndMT has been confirmedthat occur in various pathologic conditions. Transforming growth factor β1 (TGF-β1) is a potent stimulator of the vascular endothelial to mesenchymal transition (EMT). Aspirin-triggered resolvin D1 (ATRvD1) has been known to be involved in the resolution of inflammation,but whether it has effects on TGF-β1-induced EndMT is not yet clear. Therefore, we investigated the effects of AT-RvD1 on the EndMT of human umbilical vein vascular endothelial cells line (HUVECs). Treatment with TGF-β1 reduced the expression of Nrf2 and enhanced the level of F-actin, which is associated with paracellular permeability. The expression of endothelial marker VE-cadherin in HUVEC cells was reduced, and the expression of mesenchymal marker vimentin was enhanced. AT-RvD1 restored the expression of Nrf2 and vimentin and enhanced the expression of VE-cadherin. AT-RvD1 did also affect the migration of HUVEC cells. Inhibitory κB kinase 16 (IKK 16), which is known to inhibit the NF-kB pathway, had an ability to increase the expression of Nrf2 and was associated with the inhibition effect of AT-RvD1 on TGF-β1-induced EndMT, but it had no effect on TGF-β1-induced EndMT alone. Smad7, which is a key regulator of TGF-β/Smads signaling by negative feedback loops, was significantlyincreased with the treatment of AT-RvD1. These results suggest the possibility that AT-RvD1 suppresses the TGF-β1-induced EndMT through increasing the expression of Smad7 and is closely related to oxidative stress.
Collapse
Affiliation(s)
- Yusheng Shu
- Department of Cardiothoracic Surgery, Clinical Medicine College of Yangzhou University, Subei People's Hospital, Yangzhou 225001, Jiangsu, China
| | - Yu Liu
- Department of Cardiothoracic Surgery, Clinical Medicine College of Yangzhou University, Subei People's Hospital, Yangzhou 225001, Jiangsu, China
| | - Xinxin Li
- Department of Cardiothoracic Surgery, Subei People's Hospital, Yangzhou 225001, Jiangsu, China
| | - Ling Cao
- Department of Endocrinology, Clinical Medicine College of Yangzhou University, Subei People's Hospital, Yangzhou 225001, Jiangsu, China
| | - Xiaolong Yuan
- Department of Cardiothoracic Surgery, Clinical Medicine College of Yangzhou University, Subei People's Hospital, Yangzhou 225001, Jiangsu, China
| | - Wenhui Li
- Department of Cardiothoracic Surgery, Clinical Medicine College of Yangzhou University, Subei People's Hospital, Yangzhou 225001, Jiangsu, China
| | - Qianqian Cao
- Department of Cardiothoracic Surgery, Subei People's Hospital, Yangzhou 225001, Jiangsu, China
| |
Collapse
|
20
|
Rizzo AN, Sammani S, Esquinca AE, Jacobson JR, Garcia JGN, Letsiou E, Dudek SM. Imatinib attenuates inflammation and vascular leak in a clinically relevant two-hit model of acute lung injury. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1294-304. [PMID: 26432864 DOI: 10.1152/ajplung.00031.2015] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 09/27/2015] [Indexed: 12/29/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS), an illness characterized by life-threatening vascular leak, is a significant cause of morbidity and mortality in critically ill patients. Recent preclinical studies and clinical observations have suggested a potential role for the chemotherapeutic agent imatinib in restoring vascular integrity. Our prior work demonstrates differential effects of imatinib in mouse models of ALI, namely attenuation of LPS-induced lung injury but exacerbation of ventilator-induced lung injury (VILI). Because of the critical role of mechanical ventilation in the care of patients with ARDS, in the present study we pursued an assessment of the effectiveness of imatinib in a "two-hit" model of ALI caused by combined LPS and VILI. Imatinib significantly decreased bronchoalveolar lavage protein, total cells, neutrophils, and TNF-α levels in mice exposed to LPS plus VILI, indicating that it attenuates ALI in this clinically relevant model. In subsequent experiments focusing on its protective role in LPS-induced lung injury, imatinib attenuated ALI when given 4 h after LPS, suggesting potential therapeutic effectiveness when given after the onset of injury. Mechanistic studies in mouse lung tissue and human lung endothelial cells revealed that imatinib inhibits LPS-induced NF-κB expression and activation. Overall, these results further characterize the therapeutic potential of imatinib against inflammatory vascular leak.
Collapse
Affiliation(s)
- Alicia N Rizzo
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois; University of Illinois at Chicago, Department of Pharmacology, Chicago, Illinois
| | - Saad Sammani
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Adilene E Esquinca
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Jeffrey R Jacobson
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Joe G N Garcia
- Arizona Health Sciences Center, University of Arizona, Tucson, Arizona
| | - Eleftheria Letsiou
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Steven M Dudek
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois; University of Illinois at Chicago, Department of Pharmacology, Chicago, Illinois;
| |
Collapse
|
21
|
Wang L, Zhao B, Chen Y, Ma L, Chen EZ, Mao EQ. Biliary tract external drainage increases the expression levels of heme oxygenase-1 in rat livers. Eur J Med Res 2015; 20:61. [PMID: 26199001 PMCID: PMC4511237 DOI: 10.1186/s40001-015-0152-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 06/30/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Heme oxygenase-1 (HO-1) protects cells by anti-oxidation, maintaining normal microcirculation and anti-inflammatory under stress. This study investigated the effects of biliary tract external drainage (BTED) on the expression levels of HO-1 in rat livers. METHODS Biliary tract external drainage was performed by inserting a cannula into the bile duct. Sixty Sprague-Dawley rats were randomized to the following groups: sham 1 h group; BTED 1 h group; bile duct ligation (BDL) 1 h group; sham 6 h group and BTED 6 h group. The expression levels of HO-1 mRNA were analyzed using real-time RT-PCR. The expression levels of HO-1 were analyzed using immunohistochemistry. RESULTS The expression levels of HO-1 mRNA in the liver of the BTED group increased significantly compared with the sham group 1 and 6 h after surgery (p < 0.05).The expression levels of HO-1 in the BTED group increased significantly compared with the sham group 1 and 6 h after surgery. The expression levels of HO-1 mRNA in the liver in the BDL group decreased significantly compared with the sham group 1 h after surgery (p < 0.05).The expression levels of HO-1 in the BDL group decreased significantly compared with the sham group at this time. CONCLUSION Biliary tract external drainages increase the expression levels of HO-1 in the liver.
Collapse
Affiliation(s)
- Lu Wang
- Department of Emergency Intensive Care Unit, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Bing Zhao
- Department of Emergency Intensive Care Unit, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Ying Chen
- Department of Emergency Intensive Care Unit, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Li Ma
- Department of Emergency Intensive Care Unit, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Er-Zhen Chen
- Department of Emergency Intensive Care Unit, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - En-Qiang Mao
- Department of Emergency Intensive Care Unit, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
22
|
Sordi R, Chiazza F, Johnson FL, Patel NSA, Brohi K, Collino M, Thiemermann C. Inhibition of IκB Kinase Attenuates the Organ Injury and Dysfunction Associated with Hemorrhagic Shock. Mol Med 2015; 21:563-75. [PMID: 26101953 DOI: 10.2119/molmed.2015.00049] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022] Open
Abstract
Nuclear factor-kappa B (NF-κB) activation is widely implicated in multiple organ failure (MOF); however, a direct inhibitor of IκB kinase (IKK), which plays a pivotal role in the activation of NF-κB, has not been investigated in shock. Thus, the aim of the present work was to investigate the effects of an IKK inhibitor on the MOF associated with hemorrhagic shock (HS). Therefore, rats were subjected to HS and were resuscitated with the shed blood. Rats were treated with the inhibitor of IKK or vehicle at resuscitation. Four hours later, blood and organs were assessed for organ injury and signaling events involved in the activation of NF-κB. Additionally, survival following serum deprivation was assessed in HK-2 cells treated with the inhibitor of IKK. HS resulted in renal dysfunction, lung, liver and muscular injury, and increases in serum inflammatory cytokines. Kidney and liver tissue from HS rats revealed increases in phosphorylation of IKKαβ and IκBα, nuclear translocation of NF-κB and expression of inducible isoform of nitric oxide synthase (iNOS). IKK16 treatment upon resuscitation attenuated NF-κB activation and activated the Akt survival pathway, leading to a significant attenuation of all of the above parameters. Furthermore, IKK16 exhibited cytoprotective effects in human kidney cells. In conclusion, the inhibitor of IKK complex attenuated the MOF associated with HS. This effect may be due to the inhibition of the NF-κB pathway and activation of the survival kinase Akt. Thus, the inhibition of the IKK complex might be an effective strategy for the prevention of MOF associated with HS.
Collapse
Affiliation(s)
- Regina Sordi
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom.,Capes Foundation, Ministry of Education of Brazil, Brasilia, DF, Brazil
| | - Fausto Chiazza
- University of Turin, Department of Drug Science and Technology, Turin, Italy
| | - Florence L Johnson
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nimesh S A Patel
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Karim Brohi
- Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Massimo Collino
- University of Turin, Department of Drug Science and Technology, Turin, Italy
| | - Christoph Thiemermann
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
23
|
Sun X, Chen E, Dong R, Chen W, Hu Y. Nuclear factor (NF)-κB p65 regulates differentiation of human and mouse lung fibroblasts mediated by TGF-β. Life Sci 2015; 122:8-14. [DOI: 10.1016/j.lfs.2014.11.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/03/2014] [Accepted: 11/21/2014] [Indexed: 12/20/2022]
|