1
|
Zhang L, Cui H, Hu W, Meng X, Zhang C. Targeting MAD2B as a strategy for ischemic stroke therapy. J Adv Res 2024:S2090-1232(24)00269-8. [PMID: 38972542 DOI: 10.1016/j.jare.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/28/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
INTRODUCTION Post-stroke cognitive impairment is one of the major causes of disability due to cerebral ischemia. MAD2B is an inhibitor of Cdh1/APC, and loss of Cdh1/APC function in mature neurons increases ROCK2 activity, leading to changes in synaptic plasticity and memory loss in mouse neurons. Whether MAD2B regulates learning memory capacity through ROCK2 in cerebral ischemia is not known. OBJECTIVES We investigated the role and mechanism of MAD2B in cerebral ischemia-induced cognitive dysfunction. METHODS The expression of MAD2B and its downstream related molecules was detected by immunoblotting and intervened with neuroprotectants after middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation/reoxygenation (OGD/R). We constructed MAD2B-cKO-specific knockout mice, knocked down and overexpressed MAD2B in mouse hippocampus by lentiviral injection in brain stereotaxis, modeled cerebral ischemia by using MCAO, and explored the role of MAD2B in post-stroke cognitive impairment (PSCI) by animal behaviors such as Y-maze and Novel object recognition test. Then the expression of MAD2B/ROCK2, downstream molecules and apoptosis-related molecules was detected. Finally, ROCK2 expression was intervened using its inhibitor and shRNA-ROCK2 lentivirus. RESULTS The expression of MAD2B and its downstream molecules increased after MCAO and OGD/R. Nonetheless, this expression underwent a decline post-therapy with neuroprotective agents. Deletion of MAD2B in the hippocampus ameliorated memory and learning deficits and improved motor coordination in MCAO mice. Conversely, the overexpression of MAD2B in the hippocampus exacerbated learning and memory deficits. Deletion of MAD2B resulted in the downregulation of ROCK2/LIMK1/cofilin. It effectively reduced ischemia-induced upregulation of BAX and cleaved caspase-3, which could be reversed by MAD2B overexpression. Inhibition or knockdown of ROCK2 expression in primary cultured neurons led to the downregulation of LIMK1/cofilin expression and reduced the expression of apoptosis-associated molecules induced by ischemia. CONCLUSIONS Our findings suggest that MAD2B affects neuronal apoptosis via Rock2, which affects neurological function and cerebral infarction.
Collapse
Affiliation(s)
- Lijing Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hengzhen Cui
- Department of Neurobiology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wandi Hu
- Department of Neurobiology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xianfang Meng
- Department of Neurobiology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
2
|
Watanabe M, Sato T, Umetsu A, Ogawa T, Nishikiori N, Suzuki M, Furuhashi M, Ohguro H. The Specific ROCK2 Inhibitor KD025 Alleviates Glycolysis through Modulating STAT3-, CSTA- and S1PR3-Linked Signaling in Human Trabecular Meshwork Cells. Biomedicines 2024; 12:1165. [PMID: 38927372 PMCID: PMC11200618 DOI: 10.3390/biomedicines12061165] [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: 04/22/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
To investigate the biological significance of Rho-associated coiled-coil-containing protein kinase (ROCK) 2 in the human trabecular meshwork (HTM), changes in both metabolic phenotype and gene expression patterns against a specific ROCK2 inhibitor KD025 were assessed in planar-cultured HTM cells. A seahorse real-time ATP rate assay revealed that administration of KD025 significantly suppressed glycolytic ATP production rate and increased mitochondrial ATP production rate in HTM cells. RNA sequencing analysis revealed that 380 down-regulated and 602 up-regulated differentially expressed genes (DEGs) were identified in HTM cells treated with KD025 compared with those that were untreated. Gene ontology analysis revealed that DEGs were more frequently related to the plasma membrane, extracellular components and integral cellular components among cellular components, and related to signaling receptor binding and activity and protein heterodimerization activity among molecular functions. Ingenuity Pathway Analysis (IPA) revealed that the detected DEGs were associated with basic cellular biological and physiological properties, including cellular movement, development, growth, proliferation, signaling and interaction, all of which are associated with cellular metabolism. Furthermore, the upstream regulator analysis and causal network analysis estimated IL-6, STAT3, CSTA and S1PR3 as possible regulators. Current findings herein indicate that ROCK2 mediates the IL-6/STAT3-, CSTA- and S1PR3-linked signaling related to basic biological activities such as glycolysis in HTM cells.
Collapse
Affiliation(s)
- Megumi Watanabe
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (A.U.); (N.N.); (M.S.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Araya Umetsu
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (A.U.); (N.N.); (M.S.)
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Nami Nishikiori
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (A.U.); (N.N.); (M.S.)
| | - Megumi Suzuki
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (A.U.); (N.N.); (M.S.)
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, School of Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (M.W.); (A.U.); (N.N.); (M.S.)
| |
Collapse
|
3
|
Guanghui H, Zhimeng L, Yina S, Chenghua L. Nitric oxide synthase regulates coelomocytes apoptosis through the NF-κB signaling pathway in the sea cucumber Apostichopus japonicus. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109027. [PMID: 37633344 DOI: 10.1016/j.fsi.2023.109027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/28/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Nitric oxide synthase (NOS) was initially discovered to participate in the generation of nitric oxide as a defense mechanism against pathogenic infections. In recent years, it has been found that NOS plays a pivotal role in regulating apoptosis and inflammation in mammals. However, the mechanisms underlying NOS-mediated apoptosis in invertebrates remain largely unclear. In this study, we found that the Apostichopus japonicus NOS (AjNOS) expression levels were upregulated by 2.20-fold and 3.46-fold after being challenged with Vibrio splendidus at concentrations of 107 CFU mL-1 and 108 CFU mL-1 for 12 h compared to the control group, respectively. Under these conditions, the rates of coelomocytes apoptosis were increased from 14.7% to 32.7% and 45.4%, respectively. Treatment with NOS inhibitor (l-NAME) resulted in a reduction of coelomocytes apoptosis rates from 32.6% to 26.5% in V. splendidus (107 CFU mL-1) groups and from 42.3% to 33.3% in V. splendidus (108 CFU mL-1) groups, respectively. NOS has been reported to regulate apoptosis through IκBα phosphorylation. Simultaneously, exposure to V. splendidus in conjunction with l-NAME resulted in down-regulation of AjIκBα phosphorylation levels compared to the group infected solely with V. splendidus. Furthermore, immunofluorescence analysis revealed that treatment with l-NAME or interference of AjNOS using siRNA inhibited translocation of AjNF-κB/p65 (RelA) into the nucleus. Previous studies have shown that NF-κB can down-regulate expression levels of Bcl-2 family members, which is an important pathway for regulating apoptosis. In the present study, treatment with l-NAME was found to promote anti-apoptotic AjBcl-2 mRNA increase to 1.41-fold and protein expression increase to 1.86-fold at 12 h post V. splendidus challenge. However, these effects were suppressed by PMA (an NF-κB activator). Overall, our findings demonstrate that AjNOS regulates coelomocytes apoptosis induced by V. splendidus through activation of the AjNF-κB signaling pathway and down-regulation of AjBcl-2 in A. japonicus.
Collapse
Affiliation(s)
- Han Guanghui
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Lv Zhimeng
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Shao Yina
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Li Chenghua
- State-Province Joint Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, 315211, China.
| |
Collapse
|
4
|
Xue J, Zhang Z, Sun Y, Jin D, Guo L, Li X, Zhao D, Feng X, Qi W, Zhu H. Research Progress and Molecular Mechanisms of Endothelial Cells Inflammation in Vascular-Related Diseases. J Inflamm Res 2023; 16:3593-3617. [PMID: 37641702 PMCID: PMC10460614 DOI: 10.2147/jir.s418166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Endothelial cells (ECs) are widely distributed inside the vascular network, forming a vital barrier between the bloodstream and the walls of blood vessels. These versatile cells serve myriad functions, including the regulation of vascular tension and the management of hemostasis and thrombosis. Inflammation constitutes a cascade of biological responses incited by biological, chemical, or physical stimuli. While inflammation is inherently a protective mechanism, dysregulated inflammation can precipitate a host of vascular pathologies. ECs play a critical role in the genesis and progression of vascular inflammation, which has been implicated in the etiology of numerous vascular disorders, such as atherosclerosis, cardiovascular diseases, respiratory diseases, diabetes mellitus, and sepsis. Upon activation, ECs secrete potent inflammatory mediators that elicit both innate and adaptive immune reactions, culminating in inflammation. To date, no comprehensive and nuanced account of the research progress concerning ECs and inflammation in vascular-related maladies exists. Consequently, this review endeavors to synthesize the contributions of ECs to inflammatory processes, delineate the molecular signaling pathways involved in regulation, and categorize and consolidate the various models and treatment strategies for vascular-related diseases. It is our aspiration that this review furnishes cogent experimental evidence supporting the established link between endothelial inflammation and vascular-related pathologies, offers a theoretical foundation for clinical investigations, and imparts valuable insights for the development of therapeutic agents targeting these diseases.
Collapse
Affiliation(s)
- Jiaojiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yuting Sun
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Di Jin
- Department of Nephrology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Liming Guo
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiaochun Feng
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Haoyu Zhu
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| |
Collapse
|
5
|
Yang YN, Luo YB, Xu G, Li K, Ma RL, Yuan W. CircHECTD1 promoted MIRI-associated inflammation via inhibiting miR-138-5p and upregulating ROCK2. Kaohsiung J Med Sci 2023; 39:675-687. [PMID: 37096660 DOI: 10.1002/kjm2.12686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/22/2023] [Accepted: 03/10/2023] [Indexed: 04/26/2023] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) was often observed after surgeries, causing a lot of suffering to patients. Inflammation and apoptosis were critical determinants during MIRI. We conveyed experiments to reveal the regulatory functions of circHECTD1 in MIRI development. The Rat MIRI model was established and determined by 2,3,5-triphenyl tetrazolium chloride (TTC) staining. We analyzed cell apoptosis using TUNEL and flow cytometry. Proteins expression was evaluated by western blot. The RNA level was determined by qRT-PCR. Secreted inflammatory factors were analyzed by ELISA assay. To predict the interaction sequences on circHECTD1, miR-138-5p, and ROCK2, bioinformatics analysis was performed. Dual-luciferase assay was used to confirm these interaction sequences. CircHECTD1 and ROCK2 were upregulated in the rat MIRI model, while miR-138-5p was decreased. CircHECTD1 knockdown alleviated H/R-induced inflammation in H9c2 cells. Direct interaction and regulation of circHECTD1/miR-138-5p and miR-138-5p/ROCK2 were confirmed by dual-luciferase assay. CircHECTD1 promoted H/R-induced inflammation and cell apoptosis by inhibiting miR-138-5p. miR-138-5p alleviated H/R-induced inflammation, while ectopic ROCK2 antagonized such effect of miR-138-5p. Our research suggested that the circHECTD1-modulated miR-138-5p suppressing is responsible for ROCK2 activation during H/R-induced inflammatory response, providing a novel insight into MIRI-associated inflammation.
Collapse
Affiliation(s)
- Ya-Nan Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yong-Bai Luo
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Gang Xu
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kang Li
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ru-Lan Ma
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wei Yuan
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
6
|
Yan H, Wu J, Yan H. iRHOM2 regulates inflammation and endothelial barrier permeability via CX3CL1. Exp Ther Med 2023; 26:319. [PMID: 37273752 PMCID: PMC10236134 DOI: 10.3892/etm.2023.12018] [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: 09/14/2022] [Accepted: 02/16/2023] [Indexed: 06/06/2023] Open
Abstract
Acute lung injury (ALI) is associated with increased lung inflammation and lung permeability. The present study aimed to determine the role of inactive rhomboid-like protein 2 (iRHOM2) in ALI in lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial cell model. Human pulmonary microvascular endothelial cells (HPMVECs) were transfected with small interfering RNA targeting iRHOM2 and C-X3-C motif chemokine ligand 1 (CX3CL1) overexpression plasmids and treated with LPS. Cell viability was detected using a Cell Counting Kit-8 assay, while levels of TNFα, IL-1β, IL-6 and p65 were measured by reverse transcription-quantitative PCR and western blotting. Apoptosis levels were measured using a TUNEL assay. Endothelial barrier permeability was detected, followed by analysis of zonula occludens-1, vascular endothelial-cadherin and occludin by immunofluorescence staining or western blotting. The interaction of iRHOM2 and CX3CL1 was analyzed using an immune-coprecipitation assay. Through bioinformatics analysis, it was found that CX3CL1 was upregulated in the LPS group compared with the control. Kyoto Encyclopedia of Genes and Genomes pathway analysis demonstrated that the TNF signaling pathway affected by iRHOM2 and cytokine-cytokine receptor interaction, including CX3CL1, served a key role in ALI. HPMVECs treated with LPS exhibited a decrease in cell viability and an increase in inflammation, apoptosis and endothelial barrier permeability, while these effects were reversed by iRHOM2 silencing. However, CX3CL1 overexpression inhibited the effects of iRHOM2 silencing on LPS-treated HPMVECs. The present study demonstrated a novel role of iRHOM2 as a regulator that affects inflammation, apoptosis and endothelial barrier permeability; this was associated with CX3CL1.
Collapse
Affiliation(s)
- Huiyuan Yan
- Department of Internal Medicine, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Junsong Wu
- Medical College, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Huilian Yan
- Fenyang College, Shanxi Medical University, Taiyuan, Shanxi 030604, P.R. China
| |
Collapse
|
7
|
Han X. Inhibiting P2Y12 receptor relieves LPS-induced inflammation and endothelial dysfunction. Immun Inflamm Dis 2022; 10:e697. [PMID: 36169256 PMCID: PMC9449590 DOI: 10.1002/iid3.697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/14/2022] [Accepted: 08/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) is characterized by abnormal inflammatory response without effective therapies. P2Y12 receptor (P2Y12R) plays a vital role in inflammatory response. This study intends to explore whether P2Y12R antagonists can inhibit LPS-induced inflammatory injury of human pulmonary microvascular endothelial cells (HPMVECs) and endothelial cell dysfunction. METHODS Using a cell model of ALI, the role of P2Y12R was investigated in LPS-induced HPMVECs. The expression of P2Y12R was detected by RT-qPCR and Western blot analysis assay and TNF-α, IL-1β, and IL-6 levels were analyzed by RT-qPCR. NO levels were also analyzed through NO kit. The levels of NF-κB p65, P-IκB-α, and IκB-α, as well as p-AKT and eNOS levels were detected by Western blot analysis assay. Wound healing assay was performed to evaluate HPMVECs migration. FITC-dextran was used to evaluate endothelial cell permeability, and the analysis of adherens junction protein VE-cadherin and endothelial cell tight junction proteins ZO-1, Claudin 5 and Occludin expression was performed by RT-qPCR and Western blot analysis assay. RESULTS In vitro, LPS increased the expression levels of P2Y12R and pro-inflammatory mediators (TNF-α, IL-1β, and IL-6), followed by a decrease in HPMVECs migration. In addition, LPS led to an increase in endothelial cell permeability. P2Y12R antagonists Ticagrelor or clopidogrel treatment significantly reversed these effects of LPS. CONCLUSION The inhibitor of P2Y12R was able to decrease inflammatory response, promote migration and improve endothelial cell function and permeability, suggesting a key role of P2Y12R in ALI.
Collapse
Affiliation(s)
- Xiuxia Han
- Medical Department of Shandong University HospitalJinanShandongChina
| |
Collapse
|
8
|
Qian X, Yang L. ROCK2 knockdown alleviates LPS‑induced inflammatory injury and apoptosis of renal tubular epithelial cells via the NF‑κB/NLRP3 signaling pathway. Exp Ther Med 2022; 24:603. [PMID: 35949322 PMCID: PMC9353529 DOI: 10.3892/etm.2022.11540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/30/2022] [Indexed: 11/29/2022] Open
Abstract
Rho-associated protein kinase 2 (ROCK2) is an important regulator of the inflammatory response and has been reported to serve a role in sepsis. The present study aimed to investigate whether ROCK2 served a role in sepsis-associated acute kidney injury (S-AKI). HK-2 cells were stimulated with lipopolysaccharide (LPS) to simulate S-AKI in vitro. Subsequently, the change in ROCK2 expression levels were determined. ROCK2 in LPS-induced HK-2 cells was knocked down using short hairpin RNA-ROCK2, in the absence or presence of phorbol 12-myristate 13-acetate (PMA), an activator of NF-κB. Cell viability, cytotoxicity, inflammation and apoptosis were assessed using MTT, lactate dehydrogenase (LDH) release, reverse transcription-quantitative PCR, ELISA, TUNEL and western blotting assays. The protein expression levels of proteins involved in the NF-κB/NLR family pyrin domain containing 3 (NLRP3) signaling pathway were also assessed using western blotting. The results demonstrated that ROCK2 was upregulated in HK-2 cells upon LPS treatment. LPS also reduced cell viability, promoted LDH activity and increased TNF-α, IL-6 and IL-1β mRNA expression levels and concentrations. Apoptosis was also induced by LPS as indicated by an increase in the proportion of TUNEL-positive cells, decreased Bcl-2 protein expression levels and increased cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase protein expression levels. However, ROCK2 knockdown in LPS-induced HK-2 cells reversed cell viability damage and inhibited LDH activity, the generation of pro-inflammatory cytokines and apoptosis caused by LPS. Furthermore, ROCK2 knockdown inhibited the LPS-induced expression of phosphorylated-NF-κB p65, NLRP3, apoptosis-associated speck-like protein containing a CARD and caspase-1 p20. PMA treatment reversed all the aforementioned effects of ROCK2 knockdown on LPS-treated HK-2 cells. Therefore, ROCK2 knockdown may alleviate LPS-induced HK-2 cell injury via the inactivation of the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Xinfeng Qian
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, P.R. China
| | - Linjun Yang
- Department of Emergency, Hanzhong People's Hospital, Hanzhong, Shaanxi 723000, P.R. China
| |
Collapse
|
9
|
Mu Q, Lv K, Yu J, Chu S, Zhang L, Kong L, Zhang L, Tian Y, Jia X, Liu B, Wei Y, Yang N. Hydrogen Repairs LPS-Induced Endothelial Progenitor Cells Injury via PI3K/AKT/eNOS Pathway. Front Pharmacol 2022; 13:894812. [PMID: 35645804 PMCID: PMC9133378 DOI: 10.3389/fphar.2022.894812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/21/2022] [Indexed: 11/15/2022] Open
Abstract
Endotoxins and other harmful substances may cause an increase in permeability in endothelial cells (ECs) monolayers, as well as ECs shrinkage and death to induce lung damage. Lipopolysaccharide (LPS) can impair endothelial progenitor cells (EPCs) functions, including proliferation, migration, and tube formation. EPCs can migrate to the damaged area, differentiate into ECs, and participate in vascular repair, which improves pulmonary capillary endothelial dysfunction and maintains the integrity of the endothelial barrier. Hydrogen (H2) contributes to the repairment of lung injury and the damage of ECs. We therefore speculate that H2 protects the EPCs against LPS-induced damage, and it's mechanism will be explored. The bone marrow-derived EPCs from ICR Mice were treated with LPS to establish a damaged model. Then EPCs were incubated with H2, and treated with PI3K inhibitor LY294002 and endothelial nitric oxide synthase (eNOS) inhibitor L-NAME. MTT assay, transwell assay and tube formation assay were used to detect the proliferation, migration and angiogenesis of EPCs. The expression levels of target proteins were detected by Western blot. Results found that H2 repaired EPCs proliferation, migration and tube formation functions damaged by LPS. LY294002 and L-NAME significantly inhibited the repaired effect of H2 on LPS-induced dysfunctions of EPCs. H2 also restored levels of phosphor-AKT (p-AKT), eNOS and phosphor-eNOS (p-eNOS) suppressed by LPS. LY294002 significantly inhibited the increase of p-AKT and eNOS and p-eNOS expression exposed by H2. L-NAME significantly inhibited the increase of eNOS and p-eNOS expression induced by H2. H2 repairs the dysfunctions of EPCs induced by LPS, which is mediated by PI3K/AKT/eNOS signaling pathway.
Collapse
Affiliation(s)
- Qingjie Mu
- School of Clinical Medicine, Weifang Medical University, Weifang, China
- University of Health and Rehabilitation Sciences, Qingdao, China
| | - Kaixuan Lv
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Jielun Yu
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
- Medical Laboratory Animal Center, Weifang Medical University, Weifang, China
- Weifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular Diseases, Weifang, China
| | - Shangmin Chu
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Lichun Zhang
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Lingyu Kong
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, China
| | - Linlin Zhang
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Yan Tian
- Research Center of Translational Medicine Shanghai East Hospital, Tongji University, Shanghai, China
| | - Xiaopeng Jia
- Shandong Qilu Stem Cell Engineering Co., Jinan, China
| | - Benhong Liu
- Department of Respiratory, Dongying People's Hospital, Dongying, China
| | - Youzhen Wei
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Nana Yang
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
- Medical Laboratory Animal Center, Weifang Medical University, Weifang, China
- Weifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular Diseases, Weifang, China
| |
Collapse
|
10
|
Liu H, Pan Z, Ma X, Cui J, Gao J, Miao Q, Zhu Z, Chen X, Su S. ROCK inhibitor fasudil reduces the expression of inflammatory factors in LPS-induced rat pulmonary microvascular endothelial cells via ROS/NF-κB pathway. BMC Pharmacol Toxicol 2022; 23:24. [PMID: 35428330 PMCID: PMC9013060 DOI: 10.1186/s40360-022-00565-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 04/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background Inflammation plays a major role in the pulmonary artery hypertension (PAH) and the acute lung injury (ALI) diseases. The common feature of these complications is the dysfunction of pulmonary microvascular endothelial cells (PMVECs). Fasudil, the only Rho kinase (ROCK) inhibitor used in clinic, has been proved to be the most promising new drug for the treatment of PAH, with some anti-inflammatory activity. Therefore, in the present study, the effect of fasudil on lipopolysaccharide (LPS)-induced inflammatory injury in rat PMVECs was investigated. Methods LPS was used to make inflammatory injury model of rat PMVECs. Thereafter, the mRNA and protein expression of pro-inflammatory factors was evaluated by reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) assay respectively. Intracellular reactive oxygen species (ROS) levels were measured by the confocal laser scanning system. The activities of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and the content of malondialdehyde (MDA) were determined by using commercial kits according to the manufacturer’s instructions. Western blot assay was used to detect the protein expression of nuclear factor kappa B (NF-κB) p65. Results Fasudil effectively prevented inflammatory injury induced by LPS, which is manifested by the decrease of pro-inflammatory cytokines interleukin-6 (IL-6) and monocyte chenotactic protein-1 (MCP-1). Meanwhile, fasudil dramatically reduced the levels of ROS and MDA, and also elevated the activities of SOD and GSH-Px. Furthermore, the nuclear translocation of NF-κB p65 induced by LPS was also suppressed by fasudil. Additionally, the ROS scavengers N-Acetylcysteine (N-Ace) was also found to inhibit the nuclear translocation of NF-κB and the mRNA expression of IL-6 and MCP-1 induced by LPS, which suggested that ROS was essential for the nuclear translocation of NF-κB. Conclusions The present study revealed that fasudil reduced the expression of inflammatory factors, alleviated the inflammatory and oxidative damage induced by LPS in rat PMVECs via ROS-NF-κB signaling pathway.
Collapse
|
11
|
Zhou X, Wang X, Lu L, Deng M, Shi X. FGF21 improves LPS-induced pulmonary microvascular endothelial cell dysfunction and inflammatory response through SIRT1-mediated NF-κB deacetylation. Can J Physiol Pharmacol 2022; 100:492-499. [PMID: 35395162 DOI: 10.1139/cjpp-2021-0454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pneumonia is a common infectious disease of the respiratory system in children. It often leads to death in children by causing acute lung injury. Fibroblast growth factor 21 (FGF21) is a peptide hormone that plays important role in the regulation of energy homeostasis. This study aimed to investigate the role of FGF21 in alleviating the lipopolysaccharide (LPS)-induced human pulmonary microvascular endothelial cell (HPMEC) injury, as well as the underlying mechanism. The expression of SIRT1, NF-κB p65, Ac-NF-κB p65, apoptosis-related proteins, tight junction proteins and adhesion molecules in HPMECs. The viability and apoptosis of HPMECs was detected by CCK-8 and Tunel assays. LDH level and levels of inflammatory factors were respectively determined by assay kits. The mRNA expression of adhesion molecules in HPMECs was analyzed by RT-qPCR. As a result, SIRT1 expression was decreased and expression of NF-κB p65 and Ac-NF-κB p65 was increased in LPS-induced HPMECs, which were reversed by rFGF21. rFGF21 increased the viability and inhibited the apoptosis, inflammatory response, permeability and release of cell adhesion molecules of LPS-induced HPMECs. In addition, EX527 as SIRT1 inhibitor, could reversed the effect of rFGF21 on LPS-induced HPMECs. In conclusion, FGF21 improved LPS-induced HPMECs dysfunction and inflammatory response through SIRT1-mediated NF-κB deacetylation.
Collapse
Affiliation(s)
- Xuemei Zhou
- Affiliated Hospital of Jiangnan University, Department of Pediatrics, Wuxi, Jiangsu, China;
| | - Xinhua Wang
- The Affiliated Wuxi No.2 people's Hospital of Nanjing Medical University, Departmt of Respiratory Medicine, Wuxi, Jiangsu, China;
| | - Lidong Lu
- Affiliated Hospital of Jiangnan University, Department of Pediatrics, Wuxi, Jiangsu, China;
| | - Minchao Deng
- Affiliated Hospital of Jiangnan University, Department of Pediatrics, Wuxi, Jiangsu, China;
| | - Xinglei Shi
- The People's Hospital of Suichang County, Department of Pediatrics, Lishui, Zhejiang, China;
| |
Collapse
|
12
|
Yang W, Zhang Y, Lu D, Huang T, Yan K, Wang W, Gao J. Ramelteon protects against human pulmonary microvascular endothelial cell injury induced by lipopolysaccharide (LPS) via activating nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Bioengineered 2022; 13:1518-1529. [PMID: 34986734 PMCID: PMC8805948 DOI: 10.1080/21655979.2021.2021065] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Acute lung injury (ALI) is classified as a moderate or mild acute respiratory distress syndrome and is a prominent cause of morbidity and mortality among the critically ill population. Ramelteon is a melatonin receptor agonist with anti-inflammatory and antioxidant effects. The current study investigated the role of ramelteon in lipopolysaccharide (LPS)-induced human pulmonary microvascular endothelial cells (HPMECs) and its potential regulatory mechanisms. A CCK-8 assay was used to examine the effect of ramelteon on the viability of LPS-induced HPMECs, HPMECs treated with ML385 [a Nrf2 inhibitor] and HPMECs treated with SnPP [a HO-1 inhibitor]. The Nrf2/HO-1 signaling pathway was additionally assessed by performing Western blotting. The levels of oxidative stress and inflammatory cytokines in HPMECs were detected using kits and reverse transcription-quantitative PCR. Cell apoptosis was evaluated via TUNEL staining. Furthermore, cell permeability was assessed using a FITC-dextran fluorescent probe, ZO-1 and occludin expression was determined via Western blotting. The results demonstrated that ramelteon elevated HPMEC viability after LPS stimulation. Additionally, ramelteon markedly reduced LPS-induced oxidative stress, inflammation and apoptosis. Moreover, cell permeability was notably decreased in ramelteon-treated groups and was accompanied by upregulated ZO-1 and occludin expression. Ramelteon treatment also activated the Nrf2/HO-1 signaling pathway in LPS-induced HPMECs. Furthermore, the addition of ML385 or SnPP reversed the protective effects of ramelteon on LPS-induced oxidative stress, inflammation, apoptosis and cell dysfunction in HPMECs. Collectively, the results suggested that ramelteon alleviated LPS-induced HPMEC damage by activating the Nrf2/HO-1 signaling pathway, making it an effective treatment for ALI.
Collapse
Affiliation(s)
- Wenjun Yang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou City, Jiangsu Province, China
| | - Yang Zhang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou City, Jiangsu Province, China
| | - Dahao Lu
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou City, Jiangsu Province, China
| | - Tianfeng Huang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou City, Jiangsu Province, China
| | - Keshi Yan
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou City, Jiangsu Province, China
| | - Weiwei Wang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou City, Jiangsu Province, China
| | - Ju Gao
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou City, Jiangsu Province, China
| |
Collapse
|
13
|
Zhang Q, Wang Z, Zhu J, Peng Z, Tang C. Ferulic acid regulates miR-17/PTEN axis to inhibit LPS-induced pulmonary microvascular endothelial cells apoptosis through activation of PI3K/Akt pathway. J Toxicol Sci 2022; 47:61-69. [PMID: 35110471 DOI: 10.2131/jts.47.61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Qinqin Zhang
- Department of Critical Care Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, China
| | - Zhilan Wang
- Department of Critical Care Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, China
| | - Jinfei Zhu
- Department of pneumology, Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, China
| | - Zhili Peng
- Department of Critical Care Medicine, Rugao Hospital of Traditional Chinese Medicine, China
| | - Cheng Tang
- Department of Critical Care Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, China
| |
Collapse
|
14
|
Hikage F, Ichioka H, Watanabe M, Umetsu A, Ohguro H, Ida Y. ROCK inhibitors modulate the physical properties and adipogenesis of 3D spheroids of human orbital fibroblasts in different manners. FASEB Bioadv 2021; 3:866-872. [PMID: 34632320 PMCID: PMC8493964 DOI: 10.1096/fba.2021-00037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 01/01/2023] Open
Abstract
To elucidate the pharmacological effects of Rho-associated coiled-coil containing protein kinase inhibitors (ROCK-is), ripasudil (Rip), Y27632, and KD025, on human orbital fatty tissue, the human orbital fibroblasts (HOFs) were three-dimensional (3D) cultured for 12 days. The effects of ROCK-is on the physical properties of the 3D-cultured HOF spheroids, including their sizes and physical stiffness, their adipogenesis by lipid staining, and the mRNA expression of adipogenesis-related genes, PPARγ and AP2, and extracellular matrix (ECM) including collagen (COL) 1, 4, and 6, and fibronectin were analyzed. A significant increase in the sizes, physical stiffness, lipid staining, and mRNA expression of adipogenesis-related genes, COL4 and COL6, and a decrease in COL1 expression were observed with adipogenesis (DIF+). In the presence of ROCK-is, such DIF+-induced effects were differently modulated as follows: (1) the sizes were not affected or significantly enhanced by Rip, Y27632, or KD025, (2) the physical stiffness was significantly decreased in Rip and Y27632, but was substantially increased in KD025, (3) the lipid staining was further enhanced or significantly suppressed by Rip, Y27632, or KD025, and both PPARγ and AP2 expression were significantly downregulated or upregulated by KD025 or Rip, and (4) Rip upregulated the expression of COL4, Y27632 upregulated the expression of COL1, COL4, and COL6, and KD025 upregulated the expression of COL1 and COL4. This study indicates that ROCK-is significantly and differently modulate physical properties of the 3D HOF spheroids as well as their adipogenesis.
Collapse
Affiliation(s)
- Fumihito Hikage
- Departments of OphthalmologySapporo Medical University School of MedicineSapporoJapan
| | - Hanae Ichioka
- Departments of OphthalmologySapporo Medical University School of MedicineSapporoJapan
| | - Megumi Watanabe
- Departments of OphthalmologySapporo Medical University School of MedicineSapporoJapan
| | - Araya Umetsu
- Departments of OphthalmologySapporo Medical University School of MedicineSapporoJapan
| | - Hiroshi Ohguro
- Departments of OphthalmologySapporo Medical University School of MedicineSapporoJapan
| | - Yosuke Ida
- Departments of OphthalmologySapporo Medical University School of MedicineSapporoJapan
| |
Collapse
|
15
|
ROCK inhibitors enhance the production of large lipid-enriched 3D organoids of 3T3-L1 cells. Sci Rep 2021; 11:5479. [PMID: 33750898 PMCID: PMC7943807 DOI: 10.1038/s41598-021-84955-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/23/2021] [Indexed: 01/24/2023] Open
Abstract
Since the recent discovery of prostaglandin-associated peri-orbitopathy, a great deal of interest has developed concerning the side effects of anti-glaucoma medications toward periocular fatty tissue, especially their adipogenesis. Two- or three-dimension (2D or 3D) cultures of the 3T3-L1 cells were employed to elucidate the effects of the Rho-associated coiled-coil containing protein kinase inhibitor (ROCK-i) the anti-glaucoma drug, Ripasudil, and other ROCK-i, such as Y27632 on adipogenesis. Ultrastructure by electron microscopy and physical stiffness measurements by a micro-squeezer demonstrated the 3D organoids had essentially matured during the 7-day culture. The effects of ROCK-i on 3D organoid sizes, lipid staining, the mRNA expression of adipogenesis related genes, Pparγ, Cebpa and Leptin, and extracellular matrix (ECM) including collagen (COL) 1, 4 and 6, and fibronectin, and physical stiffness were then conducted. Upon adipogenesis, the sizes, lipid staining and mRNA expressions of adipogenesis related genes, Col 4 and Col 6 were dramatically increased, and were further enhanced by ROCK-i. Micro-squeezer analysis demonstrated that adipogenesis resulted in a marked less stiffed 3D organoid and this was further enhanced by ROCK-i. Our present study indicates that ROCK-i significantly enhanced the production of large lipid-enriched 3T3-L1 3D organoids.
Collapse
|
16
|
Igarashi N, Honjo M, Kaburaki T, Aihara M. Effects of ROCK Inhibitors on Apoptosis of Corneal Endothelial Cells in CMV-Positive Posner-Schlossman Syndrome Patients. Invest Ophthalmol Vis Sci 2021; 61:5. [PMID: 32749463 PMCID: PMC7441372 DOI: 10.1167/iovs.61.10.5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose To examine the role of aqueous tumor necrosis factor α (TNF-α)–RhoA–Rho kinase (ROCK) signaling in cytomegalovirus (CMV)-induced apoptosis and the barrier function of cultured human corneal endothelial cells (hCECs) in CMV-positive Posner–Schlossman syndrome (CMV+/PSS) patients. Methods Aqueous levels of TNF-α, IL-8, IL-10, and several other cytokines in 19 CMV+/PSS patients and 20 healthy control subjects were quantitated using a multiplex assay. The expression of active RhoA in hCECs post-CMV infection was determined using western blotting (WB). The expression levels of TNF-α and nuclear factor kappa B (NF-κB) in CMV-infected hCECs were examined by immunocytochemistry (ICC) and WB with and without ROCK inhibitors. The apoptotic rate and barrier integrity in CMV-infected hCECs were also examined. Results The expression levels of TNF-α, monocyte chemoattractant protein-1 (MCP-1), IL-8, and IL-10 were upregulated in the aqueous humor of CMV+/PSS patients, and among these upregulated cytokines aqueous TNF-α was negatively correlated with the number of corneal endothelial cells. In CMV-infected hCECs, upregulation of TNF-α and NF-κB was determined by WB and ICC. In hCECs, CMV infection induced apoptosis and significantly impaired cell–cell contacts, effects that were attenuated by treatment with a ROCK inhibitor. Conclusions Aqueous TNF-α was upregulated in CMV+/PSS patients, which may have triggered corneal endothelial cell loss. Modulation of TNF-α, including its downstream Rho–ROCK signaling, could serve as a novel treatment modality for corneal endothelial cell loss in CMV+/PSS patients.
Collapse
|
17
|
Liu L, Zhang H, Shi Y, Pan L. Prostaglandin E1 Improves Cerebral Microcirculation Through Activation of Endothelial NOS and GRPCH1. J Mol Neurosci 2020; 70:2041-2048. [PMID: 32483670 DOI: 10.1007/s12031-020-01610-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/21/2020] [Indexed: 12/26/2022]
Abstract
Endothelial dysfunction greatly contributes to microcirculation disorder. The role of prostaglandin E1 (PGE1) in cerebral microcirculation was explored in vitro. LPS (0.5 or 1 μg/ml) was added to induce injury in human brain microvascular endothelial cells (HCMEC/D3). CCK-8 was applied to check viabilities of HCMEC/D3 before and after LPS treatment. Western blot witnessed the changes in protein expressions of inflammatory cytokines, IL-6 and TNF-α. Caspase-3/7 activity was analyzed and so were the protein expressions of pro-apoptotic gene BAX and anti-apoptotic gene Bcl-2. mRNA expressions of eNOS and GTPCH1 were evaluated by RT-qPCR. After overexpressing eNOS or GTPCH1 in LPS-induced HCMEC/D3 cells, viabilities, inflammatory cytokines, caspase-3/7 activity, and apoptosis-related genes were detected. The modulation of PGE1 in eNOS and GTPCH1 production, viability, inflammation, and apoptosis was investigated. The inhibitor of eNOS or GTPCH1 was introduced to examine impacts of eNOS or GTPCH1 could have on the PGE1 function. LPS decreased cell viabilities, eNOS and GTPCH1 expression, and promoted inflammation and apoptosis in HCMEC/D3 cells. Overexpressed eNOS or GTPCH1 promoted cell viabilities and suppressed inflammation and apoptosis. PGE1 enhanced viability and decreased inflammation and apoptosis in cells treated by LPS. PGE1 activated eNOS and GTPCH1 and inhibition of eNOS or GTPCH1 led to the attenuation of the protective functions of PGE1 in LPS-induced cells. PGE1 protected HCMEC/D3 cells from injuries induced by LPS by activation of eNOS and GTPCH1, suggesting that PGE1 might be used to help maintain cerebral microcirculation in future.
Collapse
Affiliation(s)
- Lei Liu
- Department of Cardiology, Jinshan Hospital of Fudan University, No.1508, Longhang Road, Shanghai, 201508, China
| | - Hexi Zhang
- Department of Cardiology, Jinshan Hospital of Fudan University, No.1508, Longhang Road, Shanghai, 201508, China
| | - Yijun Shi
- Department of Cardiology, Jinshan Hospital of Fudan University, No.1508, Longhang Road, Shanghai, 201508, China
| | - Lijian Pan
- Department of Cardiology, Jinshan Hospital of Fudan University, No.1508, Longhang Road, Shanghai, 201508, China.
| |
Collapse
|
18
|
Yang Y, Han C, Sheng Y, Wang J, Zhou X, Li W, Guo L, Ruan S. The Mechanism of Aureusidin in Suppressing Inflammatory Response in Acute Liver Injury by Regulating MD2. Front Pharmacol 2020; 11:570776. [PMID: 33192512 PMCID: PMC7655772 DOI: 10.3389/fphar.2020.570776] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022] Open
Abstract
Objective In this study, we mainly explored the mechanism and target of the anti-inflammatory effects of Aureusidin (Aur) in acute liver injury. Methods Lipopolysaccharide (LPS) was used to induce inflammatory injury in Kupffer cells (KCs) in vitro. After Aur treatment with gradient concentration, flow cytometry, propidium iodide (PI) staining, and Hoechst 33342 staining were used to detect the apoptotic level of KCs, and an enzyme-linked immunosorbent assay (ELISA) was used to detect the expression levels of inflammatory factors, including Interleukin-1β (IL-1β), Interleukin-18 (IL-18), and tumor necrosis factor alpha (TNF-α). Western blot was used to detect the expression of toll-like receptor 4 (TLR4), myeloid differentiation protein-2 (MD2), MyD88, and p-P65. Aur was labeled with biotin, followed by a pull-down assay to detect its binding with MD2. Moreover, D-GalN/LPS was used to induce acute liver injury in mice in vitro, followed by Aur treatment by gavage. H&E staining was used to detect the pathological changes of liver tissue, an IF assay was used to detect the expression of MD2, Western blot was used to detect the expression of relevant proteins. Results Aur pretreatment could significantly inhibit LPS-induced KC injury, downregulate the apoptotic level, inhibit the expression of inflammatory factors, decrease the level of MDA, and downregulate the expression of MD2 in cells. Aur could inhibit the activation level of TLR4/MD2-NF-κB in a dose-dependent pattern, a high dose of Aur had a superior effect compared to low-dose Aur. In the case of MD2 deletion, the effects of Aur were suppressed. Additionally, pull-down and co-immunoprecipitation assays show that Aur can bind with the MD2 protein to inhibit the activation of TLR4/MD2-NF-κB. Results of mice experiments also showed that Aur could relieve liver injury, decrease the levels of ALT and AST, and simultaneously downregulate the levels of inflammatory factors in tissues and peripheral blood. Conclusion We found that Aur exerted an anti-inflammatory effect by directly targeting the MD2 protein, further inhibiting the expression of TLR4/MD2-NF-κB, thereby relieving acute liver injury. Therefore, Aur might be a potential inhibitor for MD2.
Collapse
Affiliation(s)
- Yi Yang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chenyang Han
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yongjia Sheng
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jin Wang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaohong Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Wenyan Li
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Li Guo
- Department of Center Laboratory, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Shuiliang Ruan
- Department of Center Laboratory, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
19
|
Chen H, Hu X, Li R, Liu B, Zheng X, Fang Z, Chen L, Chen W, Min L, Hu S. LncRNA THRIL aggravates sepsis-induced acute lung injury by regulating miR-424/ROCK2 axis. Mol Immunol 2020; 126:111-119. [PMID: 32818819 DOI: 10.1016/j.molimm.2020.07.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 01/04/2023]
Abstract
Here, we aimed to investigate the role of long noncoding RNA (lncRNA) THRIL in septic-induced acute lung injury. C57BL/6 mice were injected with Adenoviruses (Ad)-shTHRIL or negative control (NC) before caecal ligation and puncture (CLP) operation. MPVECs were transfected with Ad-shTHRIL or NC, followed by lipopolysaccharide (LPS) treatment. MiR-424 and Rho-associated kinase 2 (ROCK2) were predicted and verified as direct targets of THRIL and miR-424, respectively, by using dual-luciferase reporter assay. ROCK2 overexpression vector and shTHRIL were co-transfected into mouse pulmonary microvascular endothelial cells for 24 h before LPS treatment. Our results showed that THRIL was highly expressed in the lung of sepsis mice. CLP triggered severe lung injury and apoptosis in mice, which was abolished by THRIL knockdown. Moreover, CLP treatment visibly increased protein concentration, the number of total cell of neutrophils, and macrophages in bronchoalveolar lavage fluid (BALF). Besides, elevated protein levels of tumor necrosis factor-α, interleukin-1β, and interleukin-6 were observed in both lung and BALF. However, inhibition of THRIL reduced the number of inflammatory cells and the production of pro-inflammatory cytokines in sepsis mouse model. The effect of THRIL on inflammatory response and apoptosis in the lung was confirmed in sepsis cell model. Moreover, mechanistic studies have shown that THRIL up-regulated ROCK2 level through sponging miR-424. Furthermore, ROCK2 overexpression reversed the inhibitory effects of THRIL knockdown on LPS-induced inflammatory response and apoptosis. Overall, in vivo and in vitro results suggested that THRIL accelerates sepsis-induced lung injury by sponging miR-424 and further restoring ROCK2.
Collapse
Affiliation(s)
- Huibin Chen
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Xuemei Hu
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei Province, 430022, China
| | - Boyi Liu
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Xiang Zheng
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Zhicheng Fang
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Li Chen
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Wei Chen
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Li Min
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China
| | - Shengli Hu
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, 442000, China.
| |
Collapse
|
20
|
de Sousa GR, Vieira GM, das Chagas PF, Pezuk JA, Brassesco MS. Should we keep rocking? Portraits from targeting Rho kinases in cancer. Pharmacol Res 2020; 160:105093. [PMID: 32726671 DOI: 10.1016/j.phrs.2020.105093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 12/12/2022]
Abstract
Cancer targeted therapy, either alone or in combination with conventional chemotherapy, could allow the survival of patients with neoplasms currently considered incurable. In recent years, the dysregulation of the Rho-associated coiled-coil kinases (ROCK1 and ROCK2) has been associated with increased metastasis and poorer patient survival in several tumor types, and due to their essential roles in regulating the cytoskeleton, have gained popularity and progressively been researched as targets for the development of novel anti-cancer drugs. Nevertheless, in a pediatric scenario, the influence of both isoforms on prognosis remains a controversial issue. In this review, we summarize the functions of ROCKs, compile their roles in human cancer and their value as prognostic factors in both, adult and pediatric cancer. Moreover, we provide the up-to-date advances on their pharmacological inhibition in pre-clinical models and clinical trials. Alternatively, we highlight and discuss detrimental effects of ROCK inhibition provoked not only by the action on off-targets, but most importantly, by pro-survival effects on cancer stem cells, dormant cells, and circulating tumor cells, along with cell-context or microenvironment-dependent contradictory responses. Together these drawbacks represent a risk for cancer cell dissemination and metastasis after anti-ROCK intervention, a caveat that should concern scientists and clinicians.
Collapse
Affiliation(s)
| | | | | | | | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Brazil.
| |
Collapse
|
21
|
Yamazaki R, Hashimoto R, Masahara H, Sakamoto M, Maeno T. Time Course in Ocular Blood Flow and Pulse Waveform in a Case of Ocular Ischemic Syndrome with Intraocular Pressure Fluctuation. Vision (Basel) 2020; 4:vision4020031. [PMID: 32532078 PMCID: PMC7356182 DOI: 10.3390/vision4020031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 01/12/2023] Open
Abstract
We report on a 70-year-old Japanese man with complaints of worsening left visual acuity who was diagnosed with ocular ischemic syndrome (OIS) associated with internal carotid artery (ICA) stenosis. A gonioscopy examination showed rubeosis iridis and elevated intraocular pressure (IOP) in the left eye (50 mmHg) at the baseline visit. The optic nerve head (ONH) and choroidal blood flow measured by laser speckle flowgraphy (LSFG) was impaired in the left eye compared with that in the right eye. Additionally, the blowout score (BOS), which indicates the variation of the mean blur rate (MBR) during systolic and diastolic periods, was decreased in the left eye. After treatment with an injection of bevacizumab and administration of Rho-associated kinase-inhibitor ripasudil eye drops, both ocular blood flow and BOS in each vascular bed gradually increased along with IOP reduction. The visual acuity also improved. The current case demonstrated increased blood flow and decreased fluctuation of blood flow in the ONH and choroid before and after the treatment in OIS with rubeosis iridis. The LSFG technique is useful to non-invasively assess the ocular circulation and pulse waveform in OIS.
Collapse
Affiliation(s)
| | - Ryuya Hashimoto
- Correspondence: ; Tel.: +81-43-462-8811; Fax: +81-43-462-8820
| | | | | | | |
Collapse
|
22
|
Gao Z, Li Q, Zhang Y, Gao X, Li H, Yuan Z. Ripasudil alleviated the inflammation of RPE cells by targeting the miR-136-5p/ROCK/NLRP3 pathway. BMC Ophthalmol 2020; 20:134. [PMID: 32252692 PMCID: PMC7137504 DOI: 10.1186/s12886-020-01400-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/23/2020] [Indexed: 01/03/2023] Open
Abstract
Background Inflammation of RPE cells led to different kinds of eye diseases and affected the normal function of the retina. Furthermore, higher levels of ROCK1 and ROCK2 induced injury of endothelial cells and many inflammatory diseases of the eyes. Ripasudil, which was used for the treatment of glaucoma, was one kind of the inhibitor of ROCK1 and ROCK2, but whether ripasudil could relieve the LPS-induced inflammation and damage of RPE cells was not clear. Methods We used LPS to stimulate ARPE-19 cells, the RPE cell line. After that, we detected the levels of ROCK1 and ROCK2 by western-blotting after the stimulation of LPS and treatment of ripasudil. Then luciferase reporter assays were used to confirm the targeting effect of miR-136-5p on ROCK1 and ROCK2. At last, the levels of NLRP3, ASC, caspase1, IL-1β and IL-18 were detected with the western-blotting after the knockdown of miR-136-5p. Results The levels of ROCK1, ROCK2 and miR-136-5p in ARPE-19 cells were promoted after the stimulation of LPS. After the treatment of ripasudil, the expression levels of ROCK1, ROCK2 and miR-136-5p were suppressed. The expression of ROCK1 and ROCK2 was targeted and inhibited by the miR-136-5p. The levels of inflammation related proteins NLRP3, ASC, caspase1, IL-1β and IL-18 was also inhibited after the treatment of ripasudil. However, the expression of these proteins was rescued after the knockdown of miR-136-5p. Conclusion Ripasudil relieved the inflammatory injury of RPE cells by upregulating miR-136-5p, therefore inhibiting the expression of ROCK1, ROCK2, NLRP3, ASC, caspase1, IL-1β and IL-18.
Collapse
Affiliation(s)
- Zhao Gao
- Department of Vitreoretinopathy, Shanxi Eye Hospital, No.100 Fudong Street, Xinghualing district, Taiyuan city, 030001, Shanxi province, China
| | - Qiang Li
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan city, 030032, Shanxi province, China
| | - Yunda Zhang
- Department of Vitreoretinopathy, Shanxi Eye Hospital, No.100 Fudong Street, Xinghualing district, Taiyuan city, 030001, Shanxi province, China
| | - Xiaohong Gao
- Department of Vitreoretinopathy, Shanxi Eye Hospital, No.100 Fudong Street, Xinghualing district, Taiyuan city, 030001, Shanxi province, China
| | - Haiyan Li
- Department of Pathology, Shanxi Eye Hospital, Taiyuan city, 030001, Shanxi province, China
| | - Zhigang Yuan
- Department of Vitreoretinopathy, Shanxi Eye Hospital, No.100 Fudong Street, Xinghualing district, Taiyuan city, 030001, Shanxi province, China.
| |
Collapse
|
23
|
Abedi F, Hayes AW, Reiter R, Karimi G. Acute lung injury: The therapeutic role of Rho kinase inhibitors. Pharmacol Res 2020; 155:104736. [PMID: 32135249 DOI: 10.1016/j.phrs.2020.104736] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/18/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a pulmonary illness with high rates of mortality and morbidity. Rho GTPase and its downstream effector, Rho kinase (ROCK), have been demonstrated to be involved in cell adhesion, motility, and contraction which can play a role in ALI. The electronic databases of Google Scholar, Scopus, PubMed, and Web of Science were searched to obtain relevant studies regarding the role of the Rho/ROCK signaling pathway in the pathophysiology of ALI and the effects of specific Rho kinase inhibitors in prevention and treatment of ALI. Upregulation of the RhoA/ROCK signaling pathway causes an increase of inflammation, immune cell migration, apoptosis, coagulation, contraction, and cell adhesion in pulmonary endothelial cells. These effects are involved in endothelium barrier dysfunction and edema, hallmarks of ALI. These effects were significantly reversed by Rho kinase inhibitors. Rho kinase inhibition offers a promising approach in ALI [ARDS] treatment.
Collapse
Affiliation(s)
- Farshad Abedi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- University of South Florida, Tampa, FL, USA; Michigan State University, East Lansing, MI, USA
| | - Russel Reiter
- University of Texas, Health Science Center at San Antonio, Department of Cellular and Structural Biology, USA
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
24
|
He P, Guo Y, Wang J, Yan L, Feng A. Protection of ripasudil, a Rho kinase inhibitor, in lipopolysaccharides-induced acute pneumonia in mice. Am J Transl Res 2019; 11:6433-6443. [PMID: 31737195 PMCID: PMC6834510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 08/12/2019] [Indexed: 06/10/2023]
Abstract
Pneumonia is a major cause of morbidity and mortality of infectious diseases, especially in children. Ripasudil (K-115), a selective ROCK inhibitor, is a promising emerging drug against glaucoma, and reported to have anti-inflammatory activity. However, the anti-inflammatory effect of ripasudil still remains unclear in pneumonia. The goal of this study is to investigate the role and the underlying mechanism of ripasudil in pneumonia. BALB/c mice were used to establish an acute pneumonia model of mice by injection of lipopolysaccharide (LPS) intraperitoneally. Ripasudil (0.5 mg, 1 mg, 2 mg) was administrated 1 h before the induction of LPS. The histoligical change of lung tissue was evaluated by hematoxylin-eosin staining and lung wet/dry ratio. Inflammatory cytokines secretion, oxidant-antioxidant factors levels were measured. Cell apoptosis was examined using TNUEL assay. Western blot and qRT-PCR was used to determine gene expressions. Results showed that ripasudil significantly attenuated LPS-induced histological changes, reduced the production of pro-inflammatory cytokines, and alleviated LPS-induced oxidative stress in mice. LPS-induced cell apoptosis and associated protein expression changes were attenuated by ripasudil. Besides, ripasudil reduced the expression of RhoA, and decreased the activity of RhoA/ROCK signaling. Finally, the level of RhoA and eNOS from pneumonia patients exhibited negatively correlated, whereas the level of RhoA was higher while eNOS level was lower than that in the healthy control. The results of the present study indicate that ripasudil attenuate LPS-induced pneumonia in BALB/c mice by ameliorating inflammation, oxidative stress and apoptosis through inhibiting RhoA/ROCK signaling pathway. Ripasudil might be a novel and effective drug for the treatment of pneumonia.
Collapse
Affiliation(s)
- Ping He
- Department of Neonatology, The Second Affiliated Hospital of Shaanxi University of Chinese MedicineXianyang 712000, Shaanxi, China
| | - Yanzi Guo
- Department of Neonatology, The Second Affiliated Hospital of Shaanxi University of Chinese MedicineXianyang 712000, Shaanxi, China
| | - Junji Wang
- Department of Neonatology, The Second Affiliated Hospital of Shaanxi University of Chinese MedicineXianyang 712000, Shaanxi, China
| | - Lina Yan
- Department of Neonatology, The Second Affiliated Hospital of Shaanxi University of Chinese MedicineXianyang 712000, Shaanxi, China
| | - Aimin Feng
- Department of Neonatology, The First Affiliated Hospital of Shaanxi University of Chinese MedicineXianyang 712000, Shaanxi, China
| |
Collapse
|