1
|
Li X, Du YX, Yu CL, Niu N. Ion channels in macrophages: Implications for disease progression. Int Immunopharmacol 2025; 144:113628. [PMID: 39566388 DOI: 10.1016/j.intimp.2024.113628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
RATIONALE Macrophages are immune cells found throughout the body and exhibit morphological and functional diversity. Macrophages have been implicated in a wide range of diseases, including autoimmune diseases, acute liver injury, cardiovascular diseases, lung diseases and tumours. Ion channels are transmembrane glycoproteins with important functions in maintaining homeostasis in the intra- and extracellular environment and mediating signal transduction. Many studies have shown that different types of ion channels influence the role of macrophages in the development of various diseases. In recent years, studies on the role of ion channels in macrophages in immune regulation and inflammatory responses have attracted much attention. OBJECTIVE AND FINDINGS In order to gain a deeper understanding of the role of macrophage ion channels, this paper reviews the recent research progress on the role of macrophage ion channels in recent years. The aim is to explore the role of different ion channels in the regulation of macrophage function and their impact on a variety of disease processes. The most studied channels are calcium, sodium and potassium channels, most of which are located in the cell membrane. Among these, TRP channels have a more complex role in M1 and M2 macrophage types. CONCLUSION Ion channels are critical for the functional regulation of macrophages. Targeting ion channels provides new avenues for disease prevention and treatment. This review provides researchers with new ideas and introduces readers to the current state of research on ion channels in macrophages.
Collapse
Affiliation(s)
- Xu Li
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Yan-Xi Du
- School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Chun-Lei Yu
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Na Niu
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
2
|
Bhuvaneshwari S, Venkataraman K, Sankaranarayanan K. Exploring potential ion channel targets for rheumatoid arthritis: combination of network analysis and gene expression analysis. Biotechnol Appl Biochem 2024; 71:1405-1427. [PMID: 39049164 DOI: 10.1002/bab.2638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 07/27/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the synovial membrane that leads to the destruction of cartilage and bone. Currently, pharmacological targeting of ion channels is being increasingly recognized as an attractive and feasible strategy for the treatment of RA. The present work employs a network analysis approach to predict the most promising ion channel target for potential RA-treating drugs. A protein-protein interaction map was generated for 343 genes associated with inflammation in RA and ion channel genes using Search Tool for the Retrieval of Interacting Genes and visualized using Cytoscape. Based on the betweenness centrality and traffic values as key topological parameters, 17 hub nodes were identified, including FOS (9800.85), tumor necrosis factor (3654.60), TGFB1 (3305.75), and VEGFA (3052.88). The backbone network constructed with these 17 hub genes was intensely analyzed to identify the most promising ion channel target using network analyzer. Calcium permeating ion channels, especially store-operated calcium entry channels, and their associated regulatory proteins were found to highly interact with RA inflammatory hub genes. This significant ion channel target for RA identified by theoretical and statistical studies was further validated by a pilot case-control gene expression study. Experimental verification of the above findings in 75 RA cases and 25 controls showed increased ORAI1 expression. Thus, with a combination of network analysis approach and gene expression studies, we have explored potential targets for RA treatment.
Collapse
Affiliation(s)
- Sampath Bhuvaneshwari
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| | | | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| |
Collapse
|
3
|
Feske S, Colucci F, Coetzee WA. Do K ATP channels have a role in immunity? Front Immunol 2024; 15:1484971. [PMID: 39669557 PMCID: PMC11634800 DOI: 10.3389/fimmu.2024.1484971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/30/2024] [Indexed: 12/14/2024] Open
Abstract
Ion channels, exchangers and pumps are expressed ubiquitously in cells from all phyla of life. In mammals, their role is best described in excitable cells, where they regulate the initiation and propagation of action potentials. There are over 70 different types of K+ channels subunits that contribute to these processes. In non-excitable cells, K+ channels set the resting membrane potential, which in turn drives the activity of other translocators. K+ channels also help maintain cell volume, influence cell proliferation and apoptosis and regulate Ca2+ signaling, which in turn is crucial for many cellular processes, including metabolism, secretion, and gene expression. K+ channels play crucial roles in the activation, proliferation and a variety of other functions in cells of the innate and adaptive immune system. The ATP-sensitive K+ (KATP) channel has an established role in diverse cells, but its presence and function in immunity is scantly described. Public gene expression databases show that KATP channel subunits are highly expressed in NKT and NK cells, and that they are significantly upregulated after infection in CD8+ T cells and macrophages. We discuss these findings in the light of the available literature and propose a role for KATP channels in cytotoxicity of cells that are primed for a rapid immune response. Possible underlying molecular mechanisms are discussed.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
| | - Francesco Colucci
- Department of Obstetrics and Gynecology, University of Cambridge, Cambridge, United Kingdom
| | - William A. Coetzee
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Physiology & Neuroscience, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
4
|
Xu GY, Maskey M, Wu Z, Yang Q. Timed sulfonylurea modulation improves locomotor and sensory dysfunction following spinal cord injury. Front Pharmacol 2024; 15:1440198. [PMID: 39148545 PMCID: PMC11324438 DOI: 10.3389/fphar.2024.1440198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Traumatic spinal cord injury (SCI) results in immediate tissue necrosis and delayed secondary expansion of neurological damage, often resulting in lifelong paralysis, neurosensory dysfunction, and chronic pain. Progressive hemorrhagic necrosis (PHN) and excessive excitation are the main sources of secondary neural injury. Recent approaches to attenuate PHN by glibenclamide can improve locomotor function after SCI. However, use of glibenclamide can exacerbate development of SCI-induced chronic pain by inhibiting KATP channels to increase neuronal excitation and glial activation. In this study, we explored a treatment strategy involving administration of glibenclamide, which suppresses PHN, and diazoxide, which protects against neuronal excitation and inflammation, at different time intervals following spinal cord contusion. Our goal was to determine whether this combined approach enhances both sensory and motor function. Contusive SCI was induced at spinal segment T10 in adult rats. We found that KATP channels opener, diazoxide, decreased the hyperexcitability of primary sensory neurons after SCI by electrophysiology. Timed application of glibenclamide and diazoxide following contusion significantly improved locomotor function and mitigated development of SCI-induced chronic pain, as shown by behavioral evidence. Finally, we found that timed application of glibenclamide and diazoxide attenuates the inflammatory activity in the spinal cord and increases the survival of spinal matters following SCI. These preclinical studies introduce a promising potential treatment strategy to address SCI-induced dysfunction.
Collapse
Affiliation(s)
- Guo-Ying Xu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Manjit Maskey
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Zizhen Wu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Qing Yang
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
5
|
Alipour S, Kazemi T, Sadeghi MR, Heris JA, Masoumi J, Naseri B, Baghbani E, Sohrabi S, Baradaran B. Glyburide-treated human monocyte-derived dendritic cells loaded with insulin represent tolerogenic features with anti-inflammatory responses and modulate autologous T cell responses in vitro. Int Immunopharmacol 2024; 126:111230. [PMID: 37979448 DOI: 10.1016/j.intimp.2023.111230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
Tolerogenic dendritic cells (TolDCs) are attractive therapeutic options for autoimmune disorders because they suppress autologous T-cell responses. Dendritic cells (DCs) are equipped with pattern recognition receptors (PRR), including nucleotide-binding and oligomerization domain-like receptors (NLRs) such as NLRP3. Abnormal NLRP3 activation has been reported to be correlated with the occurrence of autoimmune disorders. Accordingly, we hypothesized that glyburide treatment of DCs by blocking the ATP-sensitive K+ (kATP) channels generates TolDCs by inhibiting NLRP3. Insulin was even loaded on a group of glyburide-treated mature DCs (mDCs) to investigate the antigen (Ag) loading effects on glyburide-treated mDCs' phenotypical and functional features. Consequently, T lymphocytes' mediated responses ensuing co-culture of them with control mDCs, insulin loaded and unloaded glyburide treated mDCs were evaluated to determine generated TolDCs' capacity in inhibition of T cell responses that are inducer of destruction in insulin-producing pancreatic beta cells in Type 1 Diabetes Mellitus (T1DM). Our findings indicated that glyburide generates desirable TolDCs with decreased surface expression of maturation and Ag presentation related markers and diminished level of inflammatory but increased level of anti-inflammatory cytokines, which even insulin loading demonstrated more anti-inflammatory functions. In addition, co-cultured T cells showed regulatory or T helper 2 phenotype instead of T helper 1 features. Our findings suggested that insulin-loaded and unloaded glyburide-treated DCs are promising therapeutic approaches for autoimmune patients, specifically DCs loaded with insulin for T1DM patients. However, further research is required before this technique can be applied in clinical practice.
Collapse
Affiliation(s)
- Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Interaction of Some Asymmetrical Porphyrins with U937 Cell Membranes-In Vitro and In Silico Studies. Molecules 2023; 28:molecules28041640. [PMID: 36838628 PMCID: PMC9959758 DOI: 10.3390/molecules28041640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
The aim of the present study was to assess the effects exerted in vitro by three asymmetrical porphyrins (5-(2-hydroxyphenyl)-10,15,20-tris-(4-acetoxy-3-methoxyphenyl)porphyrin, 5-(2-hydroxyphenyl)-10,15,20-tris-(4-acetoxy-3-methoxyphenyl)porphyrinatozinc(II), and 5-(2-hydroxyphenyl)-10,15,20-tris-(4-acetoxy-3-methoxyphenyl)porphyrinatocopper(II)) on the transmembrane potential and the membrane anisotropy of U937 cell lines, using bis-(1,3-dibutylbarbituric acid)trimethine oxonol (DiBAC4(3)) and 1-(4-trimethylammoniumphenyl)-6-phenyl-1,3,5-hexatriene p-toluenesulfonate (TMA-DPH), respectively, as fluorescent probes for fluorescence spectrophotometry. The results indicate the hyperpolarizing effect of porphyrins in the concentration range of 0.5, 5, and 50 μM on the membrane of human U937 monocytic cells. Moreover, the tested porphyrins were shown to increase membrane anisotropy. Altogether, the results evidence the interaction of asymmetrical porphyrins with the membrane of U937 cells, with potential consequences on cellular homeostasis. Molecular docking simulations, and Molecular mechanics Poisson-Boltzmann surface area (MM/PBSA) free energy of binding calculations, supported the hypothesis that the investigated porphyrinic compounds could potentially bind to membrane proteins, with a critical role in regulating the transmembrane potential. Thus, both the free base porphyrins and the metalloporphyrins could bind to the SERCA2b (sarco/endoplasmic reticulum ATPase isoform 2b) calcium pump, while the metal complexes may specifically interact and modulate calcium-dependent (large conductance calcium-activated potassium channel, Slo1/KCa1.1), and ATP-sensitive (KATP), potassium channels. Further studies are required to investigate these interactions and their impact on cellular homeostasis and functionality.
Collapse
|
7
|
Man Q, Gao Z, Chen K. Functional Potassium Channels in Macrophages. J Membr Biol 2023; 256:175-187. [PMID: 36622407 DOI: 10.1007/s00232-022-00276-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 12/30/2022] [Indexed: 01/10/2023]
Abstract
Macrophages are the predominant component of innate immunity, which is an important protective barrier of our body. Macrophages are present in all organs and tissues of the body, their main functions include immune surveillance, bacterial killing, tissue remodeling and repair, and clearance of cell debris. In addition, macrophages can present antigens to T cells and facilitate inflammatory response by releasing cytokines. Macrophages are of high concern due to their crucial roles in multiple physiological processes. In recent years, new advances are emerging after great efforts have been made to explore the mechanisms of macrophage activation. Ion channel is a class of multimeric transmembrane protein that allows specific ions to go through cell membrane. The flow of ions through ion channel between inside and outside of cell membrane is required for maintaining cell morphology and intracellular signal transduction. Expressions of various ion channels in macrophages have been detected. The roles of ion channels in macrophage activation are gradually caught attention. K+ channels are the most studied channels in immune system. However, very few of published papers reviewed the studies of K+ channels on macrophages. Here, we will review the four types of K+ channels that are expressed in macrophages: voltage-gated K+ channel, calcium-activated K+ channel, inwardly rectifying K+ channel and two-pore domain K+ channel.
Collapse
Affiliation(s)
- Qiaoyan Man
- Department of Pharmacology, Ningbo University School of Medicine, A506, Wang Changlai Building818 Fenghua Rd, Ningbo, China
| | - Zhe Gao
- Ningbo Institute of Medical Sciences, 42 Yangshan Rd, Ningbo, China.
| | - Kuihao Chen
- Department of Pharmacology, Ningbo University School of Medicine, A506, Wang Changlai Building818 Fenghua Rd, Ningbo, China.
| |
Collapse
|
8
|
Russell T, Gangotia D, Barry G. Assessing the potential of repurposing ion channel inhibitors to treat emerging viral diseases and the role of this host factor in virus replication. Biomed Pharmacother 2022; 156:113850. [DOI: 10.1016/j.biopha.2022.113850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/25/2022] [Accepted: 10/06/2022] [Indexed: 12/03/2022] Open
|
9
|
Yen FS, Wei JCC, Yu TS, Hsu CY, Hsu CC, Hwu CM. Sulfonylurea Use in Patients with Type 2 Diabetes and COPD: A Nationwide Population-Based Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15013. [PMID: 36429732 PMCID: PMC9690079 DOI: 10.3390/ijerph192215013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 06/16/2023]
Abstract
We conducted this study to investigate the long-term outcomes of sulfonylurea (SU) use in patients with chronic obstructive pulmonary disease (COPD) and type 2 diabetes (T2D). We used propensity-score matching to identify 6008 pairs of SU users and nonusers from Taiwan's National Health Insurance Research Database from 1 January 2000 to 31 December 2017. Cox proportional hazard models were used to compare the risks of mortality, cardiovascular events, non-invasive positive pressure ventilation, invasive mechanical ventilation, bacterial pneumonia, lung cancer, and hypoglycemia between SU users and nonusers. In the matched cohorts, the mean follow-up time for SU users and nonusers was 6.57 and 5.48 years, respectively. Compared with nonusers, SU users showed significantly lower risks of mortality [aHR 0.53(0.48-0.58)], cardiovascular events [aHR 0.88(0.81-0.96)], non-invasive positive pressure ventilation [aHR 0.74(0.6-0.92)], invasive mechanical ventilation [aHR 0.57(0.5-0.66)], and bacterial pneumonia [aHR 0.78(0.7-0.87)]. A longer cumulative duration of SU use was associated with a lower risk of these outcomes. This nationwide cohort study demonstrated that SU use was associated with significantly lower risks of cardiovascular events, ventilation use, bacterial pneumonia, and mortality in patients with COPD and T2D. SU may be a suitable option for diabetes management in these patients.
Collapse
Affiliation(s)
- Fu-Shun Yen
- Dr. Yen’s Clinic, No. 15, Shanying Road, Gueishan District, Taoyuan 33354, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., South District, Taichung 40201, Taiwan
- Department of Medicine, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., South District, Taichung 40201, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, No. 91, Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Teng-Shun Yu
- Management Office for Health Data, China Medical University Hospital, 3F, No. 373-2, Jianxing Road, Taichung 40459, Taiwan
- College of Medicine, China Medical University, No. 91, Xueshi Road, Taichung 40202, Taiwan
| | - Chung Y. Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Chih-Cheng Hsu
- Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
- Department of Health Services Administration, China Medical University, No. 91, Hsueh-Shih Road, Taichung 40402, Taiwan
- Department of Family Medicine, Min-Sheng General Hospital, 168 ChingKuo Road, Taoyuan 33044, Taiwan
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
| | - Chii-Min Hwu
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan
- Section of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Road, Beitou District, Taipei 11217, Taiwan
| |
Collapse
|
10
|
Selezneva A, Gibb AJ, Willis D. The contribution of ion channels to shaping macrophage behaviour. Front Pharmacol 2022; 13:970234. [PMID: 36160429 PMCID: PMC9490177 DOI: 10.3389/fphar.2022.970234] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
The expanding roles of macrophages in physiological and pathophysiological mechanisms now include normal tissue homeostasis, tissue repair and regeneration, including neuronal tissue; initiation, progression, and resolution of the inflammatory response and a diverse array of anti-microbial activities. Two hallmarks of macrophage activity which appear to be fundamental to their diverse cellular functionalities are cellular plasticity and phenotypic heterogeneity. Macrophage plasticity allows these cells to take on a broad spectrum of differing cellular phenotypes in response to local and possibly previous encountered environmental signals. Cellular plasticity also contributes to tissue- and stimulus-dependent macrophage heterogeneity, which manifests itself as different macrophage phenotypes being found at different tissue locations and/or after different cell stimuli. Together, plasticity and heterogeneity align macrophage phenotypes to their required local cellular functions and prevent inappropriate activation of the cell, which could lead to pathology. To execute the appropriate function, which must be regulated at the qualitative, quantitative, spatial and temporal levels, macrophages constantly monitor intracellular and extracellular parameters to initiate and control the appropriate cell signaling cascades. The sensors and signaling mechanisms which control macrophages are the focus of a considerable amount of research. Ion channels regulate the flow of ions between cellular membranes and are critical to cell signaling mechanisms in a variety of cellular functions. It is therefore surprising that the role of ion channels in the macrophage biology has been relatively overlooked. In this review we provide a summary of ion channel research in macrophages. We begin by giving a narrative-based explanation of the membrane potential and its importance in cell biology. We then report on research implicating different ion channel families in macrophage functions. Finally, we highlight some areas of ion channel research in macrophages which need to be addressed, future possible developments in this field and therapeutic potential.
Collapse
|
11
|
Jiang C, Xie S, Yang G, Wang N. Spotlight on NLRP3 Inflammasome: Role in Pathogenesis and Therapies of Atherosclerosis. J Inflamm Res 2022; 14:7143-7172. [PMID: 34992411 PMCID: PMC8711145 DOI: 10.2147/jir.s344730] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an intricate biological response of body tissues to detrimental stimuli. Cardiovascular disease (CVD) is the leading cause of death worldwide, and inflammation is well documented to play a role in the development of CVD, especially atherosclerosis (AS). Emerging evidence suggests that activation of the NOD-like receptor (NLR) family and the pyridine-containing domain 3 (NLRP3) inflammasome is instrumental in inflammation and may result in AS. The NLRP3 inflammasome acts as a molecular platform that triggers the activation of caspase-1 and the cleavage of pro-interleukin (IL)-1β, pro-IL-18, and gasdermin D (GSDMD). The cleaved GSDMD forms pores in the cell membrane and initiates pyroptosis, inducing cell death and the discharge of intracellular pro-inflammatory factors. Hence, the NLRP3 inflammasome is a promising target for anti-inflammatory therapy against AS. In this review, we systematically summarized the current understanding of the activation mechanism of NLRP3 inflammasome, and the pathological changes in AS involving NLRP3. We also discussed potential therapeutic strategies targeting NLRP3 inflammasome to combat AS.
Collapse
Affiliation(s)
- Chunteng Jiang
- Department of Internal Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China.,Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August-University of Göttingen, Göttingen, Lower Saxony, Germany
| | - Santuan Xie
- Department of Internal Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Guang Yang
- Department of Food Nutrition and Safety, School of Public Health, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Ningning Wang
- Department of Food Nutrition and Safety, School of Public Health, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
12
|
Wang MT, Lai JH, Huang YL, Kuo FC, Wang YH, Tsai CL, Tu MY. Use of antidiabetic medications and risk of chronic obstructive pulmonary disease exacerbation requiring hospitalization: a disease risk score-matched nested case-control study. Respir Res 2020; 21:319. [PMID: 33267895 PMCID: PMC7709288 DOI: 10.1186/s12931-020-01547-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Exacerbation of chronic obstructive pulmonary disease (COPD) severely impacts the quality of life and causes high mortality and morbidity. COPD is involved with systemic and pulmonary inflammation, which may be attenuated with antidiabetic agents exerting anti-inflammatory effects. Real-world evidence is scant regarding the effects of antidiabetic agents on COPD exacerbation. Accordingly, we conducted a disease risk score (DRS)-matched nested case-control study to systemically assess the association between each class of oral hypoglycemic agents (OHAs) and risk of severe COPD exacerbation in a nationwide COPD population co-diagnosed with diabetes mellitus (DM). METHODS We enrolled 23,875 COPD patients receiving at least one OHA for management of DM by analyzing the Taiwan National Health Insurance claims database between January 1, 2000, and December 31, 2015. Cases of severe exacerbation were defined as those who had the first hospital admission for COPD. Each case was individually matched with four randomly-selected controls by cohort entry date, DRS (the estimated probability of encountering a severe COPD exacerbation), and COPD medication regimens using the incidence density sampling approach. Conditional logistic regressions were performed to estimate odds ratios (OR) of severe COPD exacerbation for each type of OHAs. RESULTS We analyzed 2700 cases of severe COPD exacerbation and 9272 corresponding controls after DRS matching. Current use of metformin versus other OHAs was associated with a 15% (adjusted OR [aOR], 0.85; 95% confidence interval [CI] 0.75-0.95) reduced risk of severe COPD exacerbation, whereas the reduced risk was not observed with other types of antidiabetic agents. When considering the duration of antidiabetic medication therapy, current use of metformin for 91-180 and 181-365 days was associated with a 28% (aOR, 0.72; 95% CI 0.58-0.89) and 37% (aOR, 0.63; 95% CI 0.51-0.77) reduced risk of severe COPD exacerbation, respectively. Similarly, 91-180 days of sulfonylureas therapy led to a 28% (aOR, 0.72; 95% CI 0.58-0.90) lower risk, and longer treatments consistently yielded 24-30% lower risks. Current use of thiazolidinediones for more than 181 days yielded an approximately 40% decreased risk. CONCLUSIONS Duration-dependent beneficial effects of current metformin, sulfonylurea, and thiazolidinedione use on severe COPD exacerbation were observed in patients with COPD and DM.
Collapse
Affiliation(s)
- Meng-Ting Wang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Jyun-Heng Lai
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Ya-Ling Huang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan.,Department of Pharmacy, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Feng-Chih Kuo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yun-Han Wang
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Chen-Liang Tsai
- Division of Pulmonary and Critical Care, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Min-Yu Tu
- Department of Health Business Administration, Meiho University, Pingtung, Taiwan. .,Aviation Physiology Research Laboratory, Kaohsiung Armed Forces General Hospital Gangshan Branch, No.1, Dayi 2nd Rd., Gangshan Dist., Kaohsiung City, 82050, Taiwan. .,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung, Taiwan. .,Department of Life Sciences, and Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
13
|
Abstract
The accumulation of triglycerides (TGs) in macrophages induces cell death, a risk factor in the pathogenesis of atherosclerosis. We had previously reported that TG-induced macrophage death is triggered by caspase-1 and -2, therefore we investigated the mechanism underlying this phenomenon. We found that potassium efflux is increased in TG-treated THP-1 macrophages and that the inhibition of potassium efflux blocks TG-induced cell death as well as caspase-1 and -2 activation. Furthermore, reducing ATP concentration (known to induce potassium efflux), restored cell viability and caspase-1 and -2 activity. The activation of pannexin-1 (a channel that releases ATP), was increased after TG treatment in THP-1 macrophages. Inhibition of pannexin-1 activity using its inhibitor, probenecid, recovered cell viability and blocked the activation of caspase-1 and -2 in TG-treated macrophages. These results suggest that TG-induced THP-1 macrophage cell death is induced via pannexin-1 activation, which increases extracellular ATP, leading to an increase in potassium efflux.
Collapse
Affiliation(s)
- Byung Chul Jung
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, United States
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
| | - Sung Hoon Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
- Department of Biomedical Laboratory Science, Korea Nazarene University, Cheonan 31172, Korea
| | - Jaewon Lim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
- Department of Biomedical Laboratory Science, College of Medical Sciences, Daegu Haany University, Gyeongsan 38610, Korea
| | - Yoon Suk Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, Wonju 26493, Korea
| |
Collapse
|
14
|
Liu C, Guan T, Lai Y, Zhu J, Kuang J, Shen Y. ATP-sensitive potassium channels gene polymorphism rs1799858 affects the risk of macro-/micro-vascular arteriosclerotic event in patients with increased low-density lipoprotein cholesterol levels. Lipids Health Dis 2020; 19:147. [PMID: 32576189 PMCID: PMC7313205 DOI: 10.1186/s12944-020-01315-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022] Open
Abstract
Background Plasma concentration of low-density lipoprotein cholesterol (LDL-C) is causally related to the risk of arteriosclerotic events. Whether ATP-sensitive potassium channels (KATP) genetic variants predict increased LDL-C concentration (≥1.8 mmol/L) and risk of macro-/micro-vascular arteriosclerotic event remain elusive. Methods A total of 320 subjects with increased LDL-C concentration (≥1.8 mmol/L) and 320 counterpart subjects (< 1.8 mmol/L) from the South China were enrolled in this study. Three KATP polymorphisms (rs1799858, rs4148671 and rs78148713) were genotyped by the Sequenom MassARRAY system. Binary logistic regression analysis was used to evaluate the association of the 3 KATP variants with increased LDL-C concentration and carotid artery stenosis (CAS) ≥50%. Two-way ANOVA was used to analyze the association of the 3 KATP variants with microalbumin in urine (MAU) and high-sensitivity C-reactive protein (HsCRP) levels. Cox proportional hazards regression analysis was used to retrospectively analyse the association of the optimal variant with the risk of new onset/recurrent acute myocardial infarction (AMI). Results Among the 3 studied KATP gene single nucleotide polymorphisms (SNPs), only rs1799858 (TT + CT genotype) was associated with elevated risk of LDL-C ≥ 1.8 mmol/L (adjusted OR = 2.25, 95% CI: 1.31–3.85, P = 0.003) and CAS ≥50% (adjusted OR = 2.80, 95% CI: 1.12–6.98, P = 0.028). KATP SNP rs1799858 was also associated with increased MAU (P = 0.013) and HsCRP (P = 0.027) levels. The follow-up for an average of 51.1-months revealed that participants carrying the T-allele (TT + CT) of rs1799858 was associated with high risk of new onset/recurrent AMI (adjusted HR = 2.90, 95% CI: 1.06–7.94, P = 0.038). Conclusion The KATP SNP rs1799858 may be an optimal genetic predisposition marker for increased LDL-C concentration (≥1.8 mmol/L) and its related macro-/micro-vascular arteriosclerotic event risk. The KATP variant rs1799858 was associated with higher risk of macro-/micro-vascular arteriosclerotic events in patients with elevated serum LDL-C levels.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, 1 Panfu road, Guangzhou, 510180, China. .,Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China.
| | - Tianwang Guan
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Yanxian Lai
- Department of Cardiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, 1 Panfu road, Guangzhou, 510180, China
| | - Jieming Zhu
- Department of Cardiology, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Jian Kuang
- Department of Cardiology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yan Shen
- Department of Cardiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| |
Collapse
|
15
|
Arita Y, Yoshinaga Y, Kaneko T, Kawahara Y, Nakamura K, Ohgi K, Arita S, Ryu T, Takase M, Sakagami R. Glyburide inhibits the bone resorption induced by traumatic occlusion in rats. J Periodontal Res 2020; 55:464-471. [PMID: 32153049 DOI: 10.1111/jre.12731] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/25/2019] [Accepted: 12/28/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To examine whether glyburide inhibits bone destruction caused by traumatic occlusion in a rat occlusal trauma model. BACKGROUND Excessive mechanical stress, such as traumatic occlusion, induces expression of IL-1β and may be involved in bone resorption. NLRP3 inflammasomes have been linked to IL-1β expression, but it is currently unclear whether glyburide, the inhibiter of NLRP3 inflammasome, suppresses occlusal trauma in rats. METHODS Male SD rats aged 7 weeks were used. In the trauma group, the occlusal surface of the maxillary first right molar was raised by attaching a metal wire to apply occlusal trauma to the mandibular first right molar. In the trauma + glyburide group, the NLRP3 inhibitor glyburide was administered orally every 24 hours from 1 day before induction of occlusal trauma. Rats were euthanized after 5 or 10 days, and the maxillary first molars were harvested with the adjacent tissues for histopathological investigation. Immunohistochemical expression of IL-1β, NLRP3, and RANKL was also assessed. RESULTS On day 5, bone resorption was significantly greater in the trauma group compared with the control group or the trauma + glyburide group, and there were significantly higher numbers of osteoclasts and cells positive for IL-1β, NLRP3, and RANKL in the trauma group. CONCLUSION In this study, glyburide inhibits bone resorption by traumatic occlusion in rats. It suggests that the NLRP3/IL-1β pathway might be associated with bone resorption induced by traumatic occlusion.
Collapse
Affiliation(s)
- Yoichi Arita
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Yasunori Yoshinaga
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan.,Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Takashi Kaneko
- Center for Oral Diseases, Fukuoka Dental College, Fukuoka, Japan
| | - Yuri Kawahara
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Keiko Nakamura
- Center for Oral Diseases, Fukuoka Dental College, Fukuoka, Japan
| | - Kimiko Ohgi
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Seiichi Arita
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Takanori Ryu
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Minoru Takase
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Ryuji Sakagami
- Section of Periodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
16
|
MPMBP down-regulates Toll-like receptor (TLR) 2 ligand-induced proinflammatory cytokine production by inhibiting NF-κB but not AP-1 activation. Int Immunopharmacol 2020; 79:106085. [DOI: 10.1016/j.intimp.2019.106085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/23/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
|
17
|
Xia P, Cao K, Hu X, Liu L, Yu D, Dong S, Du J, Xu Y, Liu B, Yang Y, Gao F, Sun X, Liu H. K ATP Channel Blocker Glibenclamide Prevents Radiation-Induced Lung Injury and Inhibits Radiation-Induced Apoptosis of Vascular Endothelial Cells by Increased Ca 2+ Influx and Subsequent PKC Activation. Radiat Res 2019; 193:171-185. [PMID: 31877256 DOI: 10.1667/rr15381.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced lung injury (RILI) is a common and severe side effect of thoracic radiotherapy, which compromises patients' quality of life. Recent studies revealed that early vascular injury, especially microvascular damage, played a central role in the development of RILI. For this reason, early vascular protection is essential for RILI therapy. The ATP-sensitive K+ (KATP) channel is an ATP-dependent K+ channel with multiple subunits. The protective role of the KATP channel in vascular injury has been demonstrated in some published studies. In this work, we investigated the effect of KATP channel on RILI. Our findings confirmed that the KATP channel blocker glibenclamide, rather than the KATP channel opener pinacidil, remitted RILI, and in particular, provided protection against radiation-induced vascular injury. Cytology experiments verified that glibenclamide enhanced cell viability, increased the potential of proliferation after irradiation and attenuated radiation-induced apoptosis. Involved mechanisms included increased Ca2+ influx and PKC activation, which were induced by glibenclamide pretreatment. In conclusion, the KATP channel blocker glibenclamide remitted RILI and inhibited the radiation-induced apoptosis of vascular endothelial cells by increased Ca2+ influx and subsequent PKC activation.
Collapse
Affiliation(s)
- Penglin Xia
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Kun Cao
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xuguang Hu
- Department of Gastrointestinal Surgery, Changhai Hospital, Shanghai, P.R. China
| | - Lei Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Danyang Yu
- Ophthalmology Department of Kunming General Hospital of Chengdu Military Area Command, Kunming, Yunnan, China
| | - Suhe Dong
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Jicong Du
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Yang Xu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Bin Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Yanyong Yang
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xuejun Sun
- Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Hu Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
18
|
Erin N, Dilmaç S, Curry A, Duymuş Ö, Tanriover G, Prodeus A, Gariepy J, Gorczynski RM. CD200 mimetic aptamer PEG-M49 markedly increases the therapeutic effects of pegylated liposomal doxorubicin in a mouse model of metastatic breast carcinoma: an effect independent of CD200 receptor 1. Cancer Immunol Immunother 2019; 69:103-114. [PMID: 31811336 DOI: 10.1007/s00262-019-02444-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022]
Abstract
We previously reported that CD200 overexpression in the host decreases progression and metastasis of the highly aggressive metastatic 4THM breast carcinoma. We have explored a possible synergistic interaction between the CD200 mimetic PEG-M49 and pegylated liposomal doxorubicin (Peg-Dox) in wild-type CD200 knockout (CD200-/-) and CD200 Receptor 1 knockout (CD200R1-/-) mice for the first time. A 4THM breast carcinoma model and three groups of BALB/c mice (wild type, CD200-/- and CD200R1-/-) were used. Five days after injection of tumor cells, mice were injected with Peg-Dox (ip, once a week) and PEG-M49 or a control aptamer (iv, every 3 days). Necropsies were performed either 12 (mid-point) or 24 (endpoint) days after injection and the extent of tumor growth, visceral metastasis and changes in the tumor-directed immune response were evaluated. PEG-M49 and Peg-Dox co-treatment induced complete tumor regression and loss of macroscopic lung metastasis in four out of seven WT mice. This synergistic anti-tumoral effect is thought to be due to Peg-M49-induced inhibition of Gr1 + CD11b + cells and Peg-Dox-induced increases in tumor-infiltrating CD8 + and CD8CD4 double-positive cells. Similar changes were observed in CD200R1-/- mice indicating that the primary effects of Peg-M49 are mediated by non-CD200R1 receptors. We also demonstrated for the first time that tumor growth, metastasis, and tumor infiltrating GR1 + CD11b + cells were markedly increased in CD200R1-/- mice, indicating an anti-inflammatory and protective role of CD200. CD200 mimetics might be a safe and effective immunomodulatory treatment in conjunction with classical chemotherapeutics for therapy of aggressive metastatic breast carcinoma.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, Immunopharmacology and Immunooncology Research unit, Akdeniz University School of Medicine, B-Blok Kat 1 Immunoloji, 07070, Antalya, Turkey.
| | - Sayra Dilmaç
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Anna Curry
- University Health Network, Toronto General Hospital, Toronto, Canada
| | - Özlem Duymuş
- Department of Medical Pharmacology, Immunopharmacology and Immunooncology Research unit, Akdeniz University School of Medicine, B-Blok Kat 1 Immunoloji, 07070, Antalya, Turkey
| | - Gamze Tanriover
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Aaron Prodeus
- Sunnybrook Cancer Centre, Toronto General Hospital, Toronto, Canada
| | - Jean Gariepy
- Sunnybrook Cancer Centre, Toronto General Hospital, Toronto, Canada
| | | |
Collapse
|
19
|
Zou Y, Zhou C, Xu H, Yu J, Ye P, Zhang H, Chen S, Zhao J, Le S, Cui J, Jiang L, Wu J, Xia J. Glibenclamide ameliorates transplant-induced arteriosclerosis and inhibits macrophage migration and MCP-1 expression. Life Sci 2019; 241:117141. [PMID: 31811853 DOI: 10.1016/j.lfs.2019.117141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/20/2019] [Accepted: 12/01/2019] [Indexed: 01/28/2023]
Abstract
AIMS Glibenclamide, a diabetes mellitus type 2 medication, has anti-inflammatory and autoimmune properties. This study investigated the effects of glibenclamide on transplant-induced arteriosclerosis as well as the underlying molecular events. METHODS Male C57Bl/6 (H-2b) and BALB/c (H-2d) mice were used for aorta transplantation. We used hematoxylin and eosin (HE) and Elastic Van Gieson (EVG) staining for histological assessment, and qRT-PCR and ELISA to measure mRNA and protein levels. Mouse peritoneal macrophages were isolated for lipopolysaccharide (LPS) stimulation and glibenclamide treatment followed by ELISA, Western blot, and Transwell assays. RESULTS Glibenclamide inhibited transplant-induced arteriosclerosis in vivo. Morphologically, glibenclamide reduced inflammatory cell accumulation and collagen deposition in the aortas. At the gene level, glibenclamide suppressed aortic cytokine mRNA levels, including interleukin-1β (IL-1β; 10.64 ± 3.19 vs. 23.77 ± 5.72; P < .05), tumor necrosis factor-α (TNF-α; 4.59 ± 0.78 vs. 13.89 ± 5.42; P < .05), and monocyte chemoattractant protein-1 (MCP-1; 202.66 ± 23.44 vs. 1172.73 ± 208.80; P < .01), while IL-1β, TNF-α, and MCP-1 levels were also reduced in the mouse sera two weeks after glibenclamide treatment (IL-1β, 39.40 ± 13.56 ng/ml vs. 78.96 ± 9.39 ng/ml; P < .01; TNF-α, 52.60 ± 13.00 ng/ml vs. 159.73 ± 6.76 ng/ml; P < .01; and MCP-1, 56.60 ± 9.07 ng/ml vs. 223.07 ± 36.28 ng/ml; P < .001). Furthermore, glibenclamide inhibited macrophage expression and secretion of inflammatory factors in vitro through suppressing activation of the nuclear factor-κB (NF-κB) pathway and MCP-1 production. CONCLUSION Glibenclamide protected against aorta transplantation-induced arteriosclerosis by reducing inflammatory factors in vivo and inhibited macrophage migration and MCP-1 production in vitro.
Collapse
Affiliation(s)
- Yanqiang Zou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Heng Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ping Ye
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei 430022, China
| | - Hao Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shanshan Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jing Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Sheng Le
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jikai Cui
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lang Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| |
Collapse
|
20
|
Dexmedetomidine attenuates the induction and reverses the progress of 6-hydroxydopamine- induced parkinsonism; involvement of KATP channels, alpha 2 adrenoceptors and anti-inflammatory mechanisms. Toxicol Appl Pharmacol 2019; 382:114743. [DOI: 10.1016/j.taap.2019.114743] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/20/2019] [Accepted: 08/30/2019] [Indexed: 01/18/2023]
|
21
|
Zhu YR, Jiang XX, Zhang DM. Critical regulation of atherosclerosis by the KCa3.1 channel and the retargeting of this therapeutic target in in-stent neoatherosclerosis. J Mol Med (Berl) 2019; 97:1219-1229. [DOI: 10.1007/s00109-019-01814-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/07/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023]
|
22
|
Chen H, Ding Y, Chen W, Feng Y, Shi G. Glibenclamide alleviates inflammation in oleic acid model of acute lung injury through NLRP3 inflammasome signaling pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1545-1554. [PMID: 31123394 PMCID: PMC6511253 DOI: 10.2147/dddt.s196040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/02/2019] [Indexed: 12/31/2022]
Abstract
Background: Pulmonary fat embolism (PFE) is one of the important causes of acute lung injury (ALI), but its pathogenesis is unclear. In recent years, it has been found that the NLRP3 inflammasome is closely related to inflammatory response. However, there are no reports about the involvement of NLRP3 in PFE- associated ALI. Glibenclamide is a kind of hypoglycaemic drug with anti-inflammatory effect. It has been reported to have the anti-inflammatory effect related to inhibiting NLRP3. Objective: To determine whether NLRP3 inflammasome was involved in ALI induced by PFE or whether glibenclamide had therapeutic effects on such lung injury, we designed this experiment. Materials and methods: The rat model of intravenous injection of oleic acid (OA) was used to simulate PFE. Rats were divided into three groups: control, OA and glibenclamide treatment group. Blood free fatty acid (FFA) concentration was determined by ACS-ACOD. Histopathological examinations were taken to assess the severity of lung injury. The expression of NLRP3 pathway and its downstream products were analyzed by IHC, WB, qPCR and ELISA. Results: Four hours after intravenous OA injection, the typical pathological manifestations of ALI accompanied by elevated levels of plasma FFAs were found. The activity of NLRP3 inflammasomes increased in OA group, too. Pretreatment with glibenclamide partly inhibited the increase in NLRP3, caspase-1 and IL-1β expression induced by OA, simultaneously attenuated the lung injury. But it has little effect on the expression of Toll-like receptor 4 (TLR4) expression in this experiment. Conclusion: NLRP3 inflammasome, one of the main components of innate immune response, involved in ALI induced by OA. Glibenclamide can alleviate this kind of ALI by inhibiting rather the NLRP3/caspase-1/IL-1β signaling pathway than the levels of FFAs or TLR4 pathway.
Collapse
Affiliation(s)
- Hong Chen
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Yongjie Ding
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Wei Chen
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Yun Feng
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| | - Guochao Shi
- Department of Respiration and Critical Care Disease, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China.,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiaotong University, Shanghai 20025, People's Republic of China
| |
Collapse
|
23
|
Xu F, Shen G, Su Z, He Z, Yuan L. Glibenclamide ameliorates the disrupted blood-brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome. Brain Behav 2019; 9:e01254. [PMID: 30859754 PMCID: PMC6456786 DOI: 10.1002/brb3.1254] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/15/2018] [Accepted: 02/09/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Glibenclamide is a widely used sulfonylurea drug prescribed to treat type II diabetes mellitus. Previous studies have demonstrated that glibenclamide has neuroprotective effects in central nervous system injury. However, the exact mechanism by which glibenclamide acts on the blood-brain barrier (BBB) after intracerebral hemorrhage (ICH) remains unclear. The purpose of this study was to validate the neuroprotective effects of glibenclamide on ICH and to explore the mechanisms underlying these effects. METHODS We investigated the effects of glibenclamide on experimental ICH using the autologous blood infusion model. Glibenclamide was administrated either immediately or 2 hr after ICH. Brain edema was quantified using the wet-dry method 3 days after injury. BBB integrity was evaluated by Evans Blue extravasation and degradation of the tight junction protein zona occludens-1 (ZO-1). mRNA levels of inflammatory cytokines were determined by quantitative polymerase chain reaction. Activation of the nucleotide-binding oligomerization domain-like receptor with a pyrin domain 3 (NLRP3) inflammasome and cell viability were also measured in cerebral microvascular endothelial b.End3 cells exposed to hemin. Neurological changes were evaluated by the Garcia score and rotarod test. RESULTS After ICH, the brain water content, Evans Blue extravasation, and inflammatory cytokines decreased significantly in the ipsilateral hemisphere of the experimental compared to the vehicle group. Glibenclamide treatment and NLRP3 knockdown significantly reduced hemin-induced activation of the NLRP3 inflammasome, release of extracellular lactate dehydrogenase, apoptosis, and loss of ZO-1 in b.End3 cells. However, NLRP3 knockdown abolished the protective effect of glibenclamide. CONCLUSION Glibenclamide maintained BBB integrity in experimental ICH by inhibiting the activation of the NLRP3 inflammasome in microvessel endothelial cells. Our findings will contribute to elucidating the pharmacological mechanism of action of glibenclamide and to developing a novel therapy for clinical ICH.
Collapse
Affiliation(s)
- Fulin Xu
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Gang Shen
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Zuopeng Su
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Zijian He
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| | - Lutao Yuan
- Department of Neurosurgery, Minhang District Central hospital, Shanghai, China
| |
Collapse
|
24
|
Yaribeygi H, Katsiki N, Butler AE, Sahebkar A. Effects of antidiabetic drugs on NLRP3 inflammasome activity, with a focus on diabetic kidneys. Drug Discov Today 2019; 24:256-262. [DOI: 10.1016/j.drudis.2018.08.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/18/2022]
|
25
|
Yaribeygi H, Atkin SL, Pirro M, Sahebkar A. A review of the anti-inflammatory properties of antidiabetic agents providing protective effects against vascular complications in diabetes. J Cell Physiol 2018; 234:8286-8294. [PMID: 30417367 DOI: 10.1002/jcp.27699] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022]
Abstract
The global prevalence of Type 2 diabetes mellitus and its associated complications are growing rapidly. Although the role of hyperglycemia is well recognized in the pathophysiology of diabetic complications, its exact underlying mechanisms are not fully understood. In this regard, accumulating evidence suggests that the role of inflammation appears pivotal, with studies showing that most diabetic complications are associated with an inflammatory response. Several classes of antidiabetic agents have been introduced for controlling glycemia, with evidence that these pharmacological agents may have modulatory effects on inflammation beyond their glucose-lowering activity. Here we review the latest evidence on the anti-inflammatory effects of commonly used antidiabetic medications and discuss the relevance of these effects on preventing diabetic complications.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Matteo Pirro
- Department of Medicine, Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Severino P, D'Amato A, Netti L, Pucci M, De Marchis M, Palmirotta R, Volterrani M, Mancone M, Fedele F. Diabetes Mellitus and Ischemic Heart Disease: The Role of Ion Channels. Int J Mol Sci 2018; 19:802. [PMID: 29534462 PMCID: PMC5877663 DOI: 10.3390/ijms19030802] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus is one the strongest risk factors for cardiovascular disease and, in particular, for ischemic heart disease (IHD). The pathophysiology of myocardial ischemia in diabetic patients is complex and not fully understood: some diabetic patients have mainly coronary stenosis obstructing blood flow to the myocardium; others present with coronary microvascular disease with an absence of plaques in the epicardial vessels. Ion channels acting in the cross-talk between the myocardial energy state and coronary blood flow may play a role in the pathophysiology of IHD in diabetic patients. In particular, some genetic variants for ATP-dependent potassium channels seem to be involved in the determinism of IHD.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Andrea D'Amato
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Lucrezia Netti
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Mariateresa Pucci
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Marialaura De Marchis
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Raffaele Palmirotta
- Department of Biomedical Sciences and Clinical Oncology Oncogenomic Research Center, 'Aldo Moro' University of Bari, 70124 Bari, Italy.
| | - Maurizio Volterrani
- Department of Cardiac Rehabilitation, IRCCS San Raffaele, 00163 Rome, Italy.
| | - Massimo Mancone
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Francesco Fedele
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
27
|
Qu J, Tao XY, Teng P, Zhang Y, Guo CL, Hu L, Qian YN, Jiang CY, Liu WT. Blocking ATP-sensitive potassium channel alleviates morphine tolerance by inhibiting HSP70-TLR4-NLRP3-mediated neuroinflammation. J Neuroinflammation 2017; 14:228. [PMID: 29178967 PMCID: PMC5702153 DOI: 10.1186/s12974-017-0997-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/07/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Long-term use of morphine induces analgesic tolerance, which limits its clinical efficacy. Evidence indicated morphine-evoked neuroinflammation mediated by toll-like receptor 4 (TLR4) - NOD-like receptor protein 3 (NLRP3) inflammasome was important for morphine tolerance. In our study, we investigated whether other existing alternative pathways caused morphine-induced activation of TLR4 in microglia. We focused on heat shock protein 70 (HSP70), a damage-associated molecular pattern (DAMP), which was released from various cells upon stimulations under the control of KATP channel and bound with TLR4-inducing inflammation. Glibenclamide, a classic KATP channel blocker, can improve neuroinflammation by inhibiting the activation of NLRP3 inflammasome. Our present study investigated the effect and possible mechanism of glibenclamide in improving morphine tolerance via its specific inhibition on the release of HSP70 and activation of NLRP3 inflammasome induced by morphine. METHODS CD-1 mice were used for tail-flick test to evaluate morphine tolerance. The microglial cell line BV-2 and neural cell line SH-SY5Y were used to investigate the pharmacological effects and the mechanism of glibenclamide on morphine-induced neuroinflammation. The activation of microglia was accessed by immunofluorescence staining. Neuroinflammation-related cytokines were measured by western blot and real-time PCR. The level of HSP70 and related signaling pathway were evaluated by western blot and immunofluorescence staining. RESULTS Morphine induced the release of HSP70 from neurons. The released HSP70 activated microglia and triggered TLR4-mediated inflammatory response, leading to the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) p65 and the activation of NLRP3 inflammasome. Moreover, anti-HSP70 neutralizing antibody partly attenuated chronic morphine tolerance. The secretion of HSP70 was under the control of MOR/AKT/KATP/ERK signal pathway. Glibenclamide as a classic KATP channel blocker markedly inhibited the release of HSP70 induced by morphine and suppressed HSP70-TLR4-NLRP3 inflammasome-mediated neuroinflammation, which consequently attenuated morphine tolerance. CONCLUSIONS Our study indicated that morphine-induced extracellular HSP70 was an alternative way for the activation of TLR4-NLRP3 in analgesic tolerance. The release of HSP70 was regulated by MOR/AKT/KATP/ERK pathway. Our study suggested a promising target, KATP channel and a new leading compound, glibenclamide, for treating morphine tolerance.
Collapse
Affiliation(s)
- Jie Qu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Xue-You Tao
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China.,Department of Anesthesiology, Yangzhou Maternal and Child Health Hospital Affiliated with Yangzhou Medical University, Yangzhou, China.,Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Teng
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Yan Zhang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China.,Research Division of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Ci-Liang Guo
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Liang Hu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Yan-Ning Qian
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chun-Yi Jiang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China.
| | - Wen-Tao Liu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China. .,Department of Pharmacy, Sir Run Run Shaw Hospital Affiliated to Nanjing Medical University, Nanjing, China.
| |
Collapse
|
28
|
A Protective Role of Glibenclamide in Inflammation-Associated Injury. Mediators Inflamm 2017; 2017:3578702. [PMID: 28740332 PMCID: PMC5504948 DOI: 10.1155/2017/3578702] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/26/2017] [Accepted: 04/06/2017] [Indexed: 12/13/2022] Open
Abstract
Glibenclamide is the most widely used sulfonylurea drug for the treatment of type 2 diabetes mellitus (DM). Recent studies have suggested that glibenclamide reduced adverse neuroinflammation and improved behavioral outcomes following central nervous system (CNS) injury. We reviewed glibenclamide's anti-inflammatory effects: abundant evidences have shown that glibenclamide exerted an anti-inflammatory effect in respiratory, digestive, urological, cardiological, and CNS diseases, as well as in ischemia-reperfusion injury. Glibenclamide might block KATP channel, Sur1-Trpm4 channel, and NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome activation, decrease the production of proinflammatory mediators (TNF-α, IL-1β, and reactive oxygen species), and suppress the accumulation of inflammatory cells. Glibenclamide's anti-inflammation warrants further investigation.
Collapse
|
29
|
Kahnert K, Lucke T, Biertz F, Lechner A, Watz H, Alter P, Bals R, Behr J, Holle R, Huber RM, Karrasch S, Stubbe B, Wacker M, Söhler S, Wouters EFM, Vogelmeier C, Jörres RA. Transfer factor for carbon monoxide in patients with COPD and diabetes: results from the German COSYCONET cohort. Respir Res 2017; 18:14. [PMID: 28086884 PMCID: PMC5237203 DOI: 10.1186/s12931-016-0499-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/30/2016] [Indexed: 03/11/2023] Open
Abstract
Background An impairment of CO diffusing capacity has been shown in diabetic patients without lung disease. We analyzed how diffusing capacity in patients with COPD is affected by the concurrent diagnosis of diabetes. Methods Data from the initial visit of the German COPD cohort COSYCONET were used for analysis. 2575 patients with complete lung function data were included, among them 358 defined as diabetics with a reported physician diagnosis of diabetes and/or specific medication. Pairwise comparisons between groups and multivariate regression models were used to identify variables predicting the CO transfer factor (TLCO%pred) and the transfer coefficient (KCO%pred). Results COPD patients with diabetes differed from those without diabetes regarding lung function, anthropometric, clinical and laboratory parameters. Moreover, gender was an important covariate. After correction for lung function, gender and body mass index (BMI), TLCO%pred did not significantly differ between patients with and without diabetes. The results for the transfer coefficient KCO were similar, demonstrating an important role of the confounding factors RV%pred, TLC%pred, ITGV%pred, FEV1%pred, FEV1/FVC, age, packyears, creatinine and BMI. There was not even a tendency towards lower values in diabetes. Conclusion The analysis of data from a COPD cohort showed no significant differences of CO transport parameters between COPD patients with and without diabetes, if BMI, gender and the reduction in lung volumes were taken into account. This result is in contrast to observations in lung-healthy subjects with diabetes and raises the question which factors, among them potential anti-inflammatory effects of anti-diabetes medication are responsible for this finding.
Collapse
Affiliation(s)
- Kathrin Kahnert
- Department of Internal Medicine V, University of Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Ziemssenstr. 1, 80336, Munich, Germany.
| | - Tanja Lucke
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Ludwig-Maximilians-Universität München, Ziemssenstr. 1, 80336, Munich, Germany
| | - Frank Biertz
- Institute for Biostatistics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Andreas Lechner
- Department of Internal Medicine IV, University of Munich, Ziemssenstr. 1, 80336, Munich, Germany
| | - Henrik Watz
- Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North, Member of the German Center for Lung Research, Woehrendamm 80, 22927, Grosshansdorf, Germany
| | - Peter Alter
- Department of Respiratory Medicine, University of Marburg, University Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Baldingerstraße, 35043, Marburg, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, Saarland University Hospital, Kirrberger Straße 1, 66424, Homburg, Germany
| | - Jürgen Behr
- Department of Internal Medicine V, University of Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Ziemssenstr. 1, 80336, Munich, Germany
| | - Rolf Holle
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München (GmbH) - German Research Center for Environmental Health, Member of the German Center for Lung Research, Comprehensive Pneumology Center Munich (CPC-M), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Rudolf M Huber
- Department of Internal Medicine V, University of Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Ziemssenstr. 1, 80336, Munich, Germany
| | - Stefan Karrasch
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Ludwig-Maximilians-Universität München, Ziemssenstr. 1, 80336, Munich, Germany.,Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Beate Stubbe
- Department of Internal Medicine B - Cardiology, Intensive Care, Pulmonary Medicine and Infectious Diseases, Scientific Division of Pneumology and Pneumological Epidemiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475, Greifswald, Germany
| | - Margarethe Wacker
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München (GmbH) - German Research Center for Environmental Health, Member of the German Center for Lung Research, Comprehensive Pneumology Center Munich (CPC-M), Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Sandra Söhler
- ASCONET Study Coordination Office, University of Marburg, Baldingerstraße, 35043, Marburg, Germany
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Center, P. Debyelaan 25, 6202 AZ, Maastricht, The Netherlands
| | - Claus Vogelmeier
- Department of Respiratory Medicine, University of Marburg, University Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Baldingerstraße, 35043, Marburg, Germany
| | - Rudolf A Jörres
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Ludwig-Maximilians-Universität München, Ziemssenstr. 1, 80336, Munich, Germany
| | | |
Collapse
|
30
|
Abstract
Current literature shows an association of diabetes and secondary complications with chronic inflammation. Evidence of these immunological changes include altered levels of cytokines and chemokines, changes in the numbers and activation states of various leukocyte populations, apoptosis, and fibrosis during diabetes. Therefore, treatment of diabetes and its complications may include pharmacological strategies to reduce inflammation. Apart from anti-inflammatory drugs, various hypoglycemic agents have also been found to reduce inflammation that could contribute to improved outcomes. Extensive studies have been carried out with thiazolidinediones (peroxisome proliferator-activated receptor-γ agonist), dipeptidyl peptidase-4 inhibitors, and metformin (AMP-activated protein kinase activator) with each of these classes of compounds showing moderate-to-strong anti-inflammatory action. Sulfonylureas and alpha glucosidase inhibitors appeared to exert modest effects, while the injectable agents, insulin and glucagon-like peptide-1 receptor agonists, may improve secondary complications due to their anti-inflammatory potential. Currently, there is a lack of clinical data on anti-inflammatory effects of sodium–glucose cotransporter type 2 inhibitors. Nevertheless, for all these glucose-lowering agents, it is essential to distinguish between anti-inflammatory effects resulting from better glucose control and effects related to intrinsic anti-inflammatory actions of the pharmacological class of compounds.
Collapse
Affiliation(s)
- Vishal Kothari
- Department of Nutrition and Dietetics, Boshell Diabetes and Metabolic Diseases Research Program, Auburn University, Auburn, AL, USA
| | - John A Galdo
- Department of Pharmacy Practice, Samford University, Birmingham, AL, USA
| | - Suresh T Mathews
- Department of Nutrition and Dietetics, Samford University, Birmingham, AL, USA
| |
Collapse
|
31
|
Abstract
Macrophages play a critical role in regulating wound healing and tissue regeneration by changing their polarization state in response to local microenvironmental stimuli. The native roles of polarized macrophages encompass biomaterials and tissue remodeling needs, yet harnessing or directing the polarization response has been largely absent as a potential strategy to exploit in regenerative medicine to date. Recent data have revealed that specific alteration of cells' resting potential (Vmem) is a powerful tool to direct proliferation and differentiation in a number of complex tissues, such as limb regeneration, craniofacial patterning and tumorigenesis. In this study, we explored the bioelectric modulation of macrophage polarization by targeting ATP sensitive potassium channels (KATP). Glibenclamide (KATP blocker) and pinacidil (KATP opener) treatment not only affect macrophage polarization, but also influence the phenotype of prepolarized macrophages. Furthermore, modulation of cell membrane electrical properties can fine-tune macrophage plasticity. Glibenclamide decreased the secretion and gene expression of selected M1 markers, while pinacidil augmented M1 markers. More interestingly, glibencalmide promoted macrophage alternative activation by enhancing certain M2 markers during M2 polarization. These findings suggest that control of bioelectric properties of macrophages could offer a promising approach to regulate macrophage phenotype as a useful tool in regenerative medicine.
Collapse
|
32
|
RamaKrishnan AM, Sankaranarayanan K. Understanding autoimmunity: The ion channel perspective. Autoimmun Rev 2016; 15:585-620. [PMID: 26854401 DOI: 10.1016/j.autrev.2016.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 01/29/2016] [Indexed: 12/11/2022]
Abstract
Ion channels are integral membrane proteins that orchestrate the passage of ions across the cell membrane and thus regulate various key physiological processes of the living system. The stringently regulated expression and function of these channels hold a pivotal role in the development and execution of various cellular functions. Malfunction of these channels results in debilitating diseases collectively termed channelopathies. In this review, we highlight the role of these proteins in the immune system with special emphasis on the development of autoimmunity. The role of ion channels in various autoimmune diseases is also listed out. This comprehensive review summarizes the ion channels that could be used as molecular targets in the development of new therapeutics against autoimmune disorders.
Collapse
Affiliation(s)
| | - Kavitha Sankaranarayanan
- AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chrompet, Chennai 600 044, India.
| |
Collapse
|
33
|
Kv1.3 potassium channel mediates macrophage migration in atherosclerosis by regulating ERK activity. Arch Biochem Biophys 2016; 591:150-6. [DOI: 10.1016/j.abb.2015.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 12/05/2015] [Accepted: 12/29/2015] [Indexed: 12/30/2022]
|
34
|
The roles of macrophage autophagy in atherosclerosis. Acta Pharmacol Sin 2016; 37:150-6. [PMID: 26750103 DOI: 10.1038/aps.2015.87] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/07/2015] [Indexed: 12/11/2022]
Abstract
Although various types of drugs and therapies are available to treat atherosclerosis, it remains a major cause of mortality throughout the world. Macrophages are the major source of foam cells, which are hallmarks of atherosclerotic lesions. Consequently, the roles of macrophages in the pathophysiology of atherosclerosis are increasingly investigated. Autophagy is a self-protecting cellular catabolic pathway. Since its discovery, autophagy has been found to be associated with a variety of diseases, including cardiovascular diseases, malignant tumors, neurodegenerative diseases, and immune system disorders. Accumulating evidence demonstrates that autophagy plays an important role in inhibiting inflammation and apoptosis, and in promoting efferocytosis and cholesterol efflux. These facts suggest the induction of autophagy may be exploited as a potential strategy for the treatment of atherosclerosis. In this review we mainly discuss the relationship between macrophage autophagy and atherosclerosis and the molecular mechanisms, as well as the recent advances in targeting the process of autophagy to treat atherosclerosis.
Collapse
|
35
|
Antidiabetic agents: Potential anti-inflammatory activity beyond glucose control. DIABETES & METABOLISM 2015; 41:183-94. [DOI: 10.1016/j.diabet.2015.02.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/09/2015] [Indexed: 12/13/2022]
|
36
|
Winkel LC, Hoogendoorn A, Xing R, Wentzel JJ, Van der Heiden K. Animal models of surgically manipulated flow velocities to study shear stress-induced atherosclerosis. Atherosclerosis 2015; 241:100-10. [PMID: 25969893 DOI: 10.1016/j.atherosclerosis.2015.04.796] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/12/2015] [Accepted: 04/22/2015] [Indexed: 10/23/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial tree that develops at predisposed sites, coinciding with locations that are exposed to low or oscillating shear stress. Manipulating flow velocity, and concomitantly shear stress, has proven adequate to promote endothelial activation and subsequent plaque formation in animals. In this article, we will give an overview of the animal models that have been designed to study the causal relationship between shear stress and atherosclerosis by surgically manipulating blood flow velocity profiles. These surgically manipulated models include arteriovenous fistulas, vascular grafts, arterial ligation, and perivascular devices. We review these models of manipulated blood flow velocity from an engineering and biological perspective, focusing on the shear stress profiles they induce and the vascular pathology that is observed.
Collapse
Affiliation(s)
- Leah C Winkel
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Ayla Hoogendoorn
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Ruoyu Xing
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jolanda J Wentzel
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Kim Van der Heiden
- Department of Biomedical Engineering, Erasmus Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
37
|
Collin J, Gössl M, Matsuo Y, Cilluffo RR, Flammer AJ, Loeffler D, Lennon RJ, Simari RD, Spoon DB, Erbel R, Lerman LO, Khosla S, Lerman A. Osteogenic monocytes within the coronary circulation and their association with plaque vulnerability in patients with early atherosclerosis. Int J Cardiol 2014; 181:57-64. [PMID: 25482280 DOI: 10.1016/j.ijcard.2014.11.156] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 09/08/2014] [Accepted: 11/23/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVES This study tests the hypothesis that circulating mononuclear cells expressing osteocalcin (OCN) and bone alkaline phosphatase (BAP) are associated with distinct plaque tissue components in patients with early coronary atherosclerosis. BACKGROUND Plaque characteristics implying vulnerability develop at the earliest stage of coronary atherosclerosis. Increasing evidence indicates that cells from the myeloid lineage might serve as important mediators of destabilization. Plaque burden and its components were assessed regarding their relationship to monocytes carrying both pro-inflammatory (CD14) and osteogenic surface markers OCN and BAP. METHODS Twenty-three patients with angiographically non-obstructive coronary artery disease underwent coronary endothelial function assessment and virtual histology-intravascular ultrasound of the left coronary artery. Plaque composition was characterized in the total segment (TS) and in the target lesion (TL) containing the highest amount of plaque burden. Blood samples were collected simultaneously from the aorta and the coronary sinus. Circulating cell counts were then identified from each sample and a gradient across the coronary circulation was determined. RESULTS Circulating CD14+/BAP+/OCN+ monocytes correlate with the extent of necrotic core and calcification (r=0.53, p=0.010; r=0.55, p=0.006, respectively). Importantly, coronary retention of CD14+/OCN+ cells also correlates with the amount of necrotic core and calcification (r=0.61, p=0.003; r=0.61, p=0.003) respectively. CONCLUSIONS Our study links CD14+/BAP+/OCN+ monocytes to the pathologic remodeling of the coronary circulation and therefore associates these cells with plaque destabilization in patients with early coronary atherosclerosis.
Collapse
Affiliation(s)
- Julia Collin
- Department of Cardiology, West-German Heart-Center - University Hospital Essen, Essen, Germany; Department of Internal Medicine, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Mario Gössl
- Department of Cardiology, Dean Clinic - Dean St. Mary's Outpatient Hospital Center, Madison, WI, USA
| | - Yoshiki Matsuo
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Rebecca R Cilluffo
- Divison of Cardiovascular Diseases and Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Andreas J Flammer
- Department of Cardiology, University Hospital of Zurich, Zurich, Switzerland
| | - Darrell Loeffler
- Divison of Cardiovascular Diseases and Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Ryan J Lennon
- Division of Biomedical Statistics and Informatics, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Robert D Simari
- Department of Internal Medicine, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Daniel B Spoon
- Divison of Cardiovascular Diseases and Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Raimund Erbel
- Department of Cardiology, West-German Heart-Center - University Hospital Essen, Essen, Germany
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Sundeep Khosla
- Division of Endocrinology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Amir Lerman
- Divison of Cardiovascular Diseases and Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA.
| |
Collapse
|
38
|
Cai J, Lu S, Yao Z, Deng YP, Zhang LD, Yu JW, Ren GF, Shen FM, Jiang GJ. Glibenclamide attenuates myocardial injury by lipopolysaccharides in streptozotocin-induced diabetic mice. Cardiovasc Diabetol 2014; 13:106. [PMID: 25077824 PMCID: PMC4147163 DOI: 10.1186/s12933-014-0106-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 01/25/2014] [Accepted: 06/22/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Sepsis is a common disease that continues to increase in incidence in the world. Diseases, such as diabetes mellitus, may make the situation worse. Diabetic patients are at increased risk for common infections. This study was designed to investigate the role of glibenclamide on myocardial injury by lipopolysaccharides (LPS) in streptozotocin induced diabetic mice (STZ-mice). METHODS LPS was used to induce endotoxemia in STZ-mice. Heart rate and mean arterial pressure were measured by MPA-HBBS. Serum epinephrine level was measured by enzyme-linked immunosorbent assays (ELISA). Myocardial injury was examined by light and transmission electron microscope and TUNEL staining. Macrophage infiltration was measured by immunohistochemistry. Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) levels in myocardial tissue and serum in STZ-mice, and in conditional medium of primary cultured peritoneal macrophages were determined by ELISA. Nalp3 and Caspase-1 protein levels were measured by Western blotting analysis. RESULTS STZ administration decreased body weight and increased blood glucose in C57BL/6 mice. LPS injection caused decreases of heart rate and mean arterial pressure, and elevated serum epinephrine level in C57BL/6 mice. Compared with control mice without STZ treatment, LPS induced more severe myocardial injury and macrophage infiltration in STZ-mice, which was attenuated by pretreatment of glibenclamide. LPS stimulation enhanced the levels of IL-1β and TNF-α in both cardiac tissue and serum. Glibenclamide pretreatment significantly inhibited the serum levels of pro-inflammatory cytokines. Either high glucose or LPS increased the levels of IL-1β and TNF-α in the conditional medium of peritoneal macrophages. Glibenclamide treatment suppressed the increase of IL-1β level induced by high glucose and LPS. Furthermore, Nalp3 and Caspase-1 levels were markedly increased by high glucose plus LPS, and both proteins were significantly inhibited by glibenclamide treatment. CONCLUSIONS We conclude that glibenclamide could attenuate myocardial injury induced by LPS challenge in STZ-mice, which was possibly related to inhibiting inflammation through Nalp3 inflammasomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fu-Ming Shen
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou 311202, Zhejiang, China.
| | | |
Collapse
|
39
|
Glibenclamide decreases ATP-induced intracellular calcium transient elevation via inhibiting reactive oxygen species and mitochondrial activity in macrophages. PLoS One 2014; 9:e89083. [PMID: 24558474 PMCID: PMC3928368 DOI: 10.1371/journal.pone.0089083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 01/19/2014] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence has revealed that glibenclamide has a wide range of anti-inflammatory effects. However, it is unclear whether glibenclamide can affect the resting and adenosine triphosphate (ATP)-induced intracellular calcium ([Ca2+]i) handling in Raw 264.7 macrophages. In the present study, [Ca2+]i transient, reactive oxygen species (ROS) and mitochondrial activity were measured by the high-speed TILLvisION digital imaging system using the indicators of Fura 2-am, DCFDA and rhodamine-123, respectively. We found that glibenclamide, pinacidil and other unselective K+ channel blockers had no effect on the resting [Ca2+]i of Raw 264.7 cells. Extracellular ATP (100 µM) induced [Ca2+]i transient elevation independent of extracellular Ca2+. The transient elevation was inhibited by an ROS scavenger (tiron) and mitochondria inhibitor (rotenone). Glibenclamide and 5-hydroxydecanoate (5-HD) also decreased ATP-induced [Ca2+]i transient elevation, but pinacidil and other unselective K+ channel blockers had no effect. Glibenclamide also decreased the peak of [Ca2+]i transient induced by extracellular thapsigargin (Tg, 1 µM). Furthermore, glibenclamide decreased intracellular ROS and mitochondrial activity. When pretreated with tiron and rotenone, glibenclamide could not decrease ATP, and Tg induced maximal [Ca2+]i transient further. We conclude that glibenclamide may inhibit ATP-induced [Ca2+]i transient elevation by blocking mitochondria KATP channels, resulting in decreased ROS generation and mitochondrial activity in Raw 264.7 macrophages.
Collapse
|