1
|
Ciftel S, Mercantepe F, Mercantepe T, Ciftel E, Klisic A. Dexmedetomidine on the interplay of IL-6 and STAT3 pathways in adrenal gland damage-induced scalding burns in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:641-655. [PMID: 39042159 PMCID: PMC11787240 DOI: 10.1007/s00210-024-03300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/14/2024] [Indexed: 07/24/2024]
Abstract
Scalding burns are a common form of thermal injury that often leads to systemic complications. Pro-inflammatory cytokines like interleukin-6 (IL-6) and the activation of signal transducer and activator of transcription 3 (STAT3) pathways have been linked to the pathophysiology of organ damage caused by burns. This study aimed to investigate the potential therapeutic effects of dexmedetomidine, an α2-adrenergic receptor agonist with anti-inflammatory properties, on the interplay of IL-6 and STAT3 pathways in adrenal gland damage following scalding burns in rats. Twenty-eight rats were divided randomly into four groups. Rats in group 1 (n=7, control) were given only 0.9% intraperitoneal (i.p.) NaCl. Rats in group 2 (n=7, DEX) were exposed to 25°C water for 17 s on day 1 and received 100 mcg/kg/day dexmedetomidine i.p. for 3 days; for rats in group 3 (n=7, Burn), boiling water of 94°C was applied inside for 17 s. Rats in group 4 (n=7, Burn+DEX) were exposed to 94°C water for 17 s and received 100 mcg/kg/day dexmedetomidine i.p. for 3 days. Adrenal gland tissues were histopathological examined, and STAT3, IL-6, and TUNEL staining were performed using immunohistochemically. Our results revealed that scalding burns increased IL-6 and STAT3 expression in the adrenal glands of rats. Histological analysis demonstrated that dexmedetomidine administration ameliorated adrenal gland damage and reduced inflammatory cell infiltration. Our findings suggest that dexmedetomidine protects the adrenal glands in scalding burns. This protection appears to be mediated, at least in part, by its modulation of IL-6 and STAT3 pathways.
Collapse
Affiliation(s)
- Serpil Ciftel
- Department of Endocrinology and Metabolism, Erzurum Regional Training and Research Hospital, Erzurum, Turkey
| | - Filiz Mercantepe
- Department of Endocrinology and Metabolism, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, 53010, Turkey.
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Enver Ciftel
- Department of Endocrinology and Metabolism, Sivas Numune Hospital, Sivas, Turkey
| | - Aleksandra Klisic
- University of Montenegro-Faculty of Medicine, Podgorica, Montenegro
- Center for Laboratory Diagnostics, Primary Health Care Center, Podgorica, Montenegro
| |
Collapse
|
2
|
Zheng ZL, Ma JW, Luo Y, Liang GJ, Lei SJ, Yan KJ, Meng HB, Liu XJ. Mechanism of dexmedetomidine protection against cisplatin induced acute kidney injury in rats. Ren Fail 2024; 46:2337287. [PMID: 38627212 PMCID: PMC11022910 DOI: 10.1080/0886022x.2024.2337287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE This study explored the molecular mechanisms by which dexmedetomidine (Dex) alleviates cisplatin (CP)-induced acute kidney injury (AKI) in rats. METHODS CP-induced AKI models were established, and Dex was intraperitoneally injected at different concentrations into rats in the model groups. Subsequently, rats were assigned to the control, CP, CP + Dex 10 μg/kg, and CP + Dex 25 μg/kg groups. After weighing the kidneys of the rats, the kidney arterial resistive index was calculated, and CP-induced AKI was evaluated. In addition, four serum biochemical indices were measured: histopathological damage in rat kidneys was detected; levels of inflammatory factors, interleukin (IL)-1β, IL-18, IL-6, and tumor necrosis factor alpha, in kidney tissue homogenate of rats were assessed through enzyme-linked immunosorbent assay (ELISA); and levels of NLRP-3, caspase-1, cleaved caspase-1, gasdermin D (GSDMD), and GSDMD-N in kidney tissues of rats were determined via western blotting. RESULTS Dex treatment reduced nephromegaly and serum clinical marker upregulation caused by CP-induced AKI. In addition, hematoxylin and eosin staining revealed that Dex treatment relieved CP-induced kidney tissue injury in AKI rats. ELISA analyses demonstrated that Dex treatment reduced the upregulated levels of proinflammatory cytokines in the kidney tissue of AKI rats induced by CP, thereby alleviating kidney tissue injury. Western blotting indicated that Dex alleviated CP-induced AKI by inhibiting pyroptosis mediated by NLRP-3 and caspase-1. CONCLUSION Dex protected rats from CP-induced AKI, and the mechanism may be related to NLRP-3/Caspase-1-mediated pyroptosis.
Collapse
Affiliation(s)
- Zeng-lu Zheng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Jun-wei Ma
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Yi Luo
- Department of Respiratory, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Gui-jin Liang
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Shi-jie Lei
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Ke-jin Yan
- Department of Proctology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Hai-bing Meng
- Department of Anesthesiology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| | - Xiu-juan Liu
- Department of Nephrology, The 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, China
| |
Collapse
|
3
|
Yavuz A, Küçük A, Ergörün Aİ, Dursun AD, Yiğman Z, Alkan M, Arslan M. Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation. Exp Ther Med 2024; 27:242. [PMID: 38655036 PMCID: PMC11036365 DOI: 10.3892/etm.2024.12530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/27/2023] [Indexed: 04/26/2024] Open
Abstract
Sepsis is a systemic inflammatory response syndrome that develops in the host against microorganisms. This response develops away from the primary infection site and results in end-organ damage. The present study aimed to investigate the protective and therapeutic effects on lung and kidney tissue of silymarin (S) and dexmedetomidine (DEX) applied 1 h before and after sepsis induced by the cecal ligation and puncture (CLP) method in rats. A total of 62 rats was randomly divided into eight groups: i) Control (n=6); ii) cecal perforation (CLP; n=8); iii) S + CLP (n=8; S + CLP; S administered 1 h before CPL); iv) CLP + S (n=8; S administered 1 h after CLP); v) DEX + CLP (n=8; D + CLP; DEX administered 1 h before CLP); vi) CLP + D (n=8; DEX administered 1 h after CLP); vii) SD + CLP (n=8; S and DEX administered 1 h before CLP) and viii) CLP + SD (n=8; S and DEX administered 1 h after CLP). After the cecum filled with stool, it was tied with 3/0 silk under the ileocecal valve and the anterior surface of the cecum was punctured twice with an 18-gauge needle. A total of 100 mg/kg silymarin and 100 µg/kg DEX were administered intraperitoneally to the treatment groups. Lung and kidney tissue samples were collected to evaluate biochemical and histopathological parameters. In the histopathological examination, all parameters indicating kidney injury; interstitial edema, peritubular capillary dilatation, vacuolization, ablation of tubular epithelium from the basement membrane, loss of brush border in the proximal tubule epithelium, cell swelling and nuclear defragmentation; were increased in the CLP compared with the control group. Silymarin administration increased kidney damage, including ablation of tubular epithelium from the basement membrane, compared with that in the CLP group. DEX significantly reduced kidney damage compared with the CLP and silymarin groups. The co-administration of DEX + silymarin decreased kidney damage, although it was not as effective as DEX-alone. To conclude, intraperitoneal DEX ameliorated injury in CLP rats. DEX + silymarin partially ameliorated injury but silymarin administration increased damage. As a result, silymarin has a negative effects with this dosage and DEX has a protective effect. In the present study, it was determined that using the two drugs together had a greater therapeutic effect than silymarin and no differences in the effects were not observed any when the application times of the agents were changed.
Collapse
Affiliation(s)
- Aydin Yavuz
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ayşegül Küçük
- Department of Physiology, Faculty of Medicine, Kutahya Health Science University, Kutahya 43020, Turkey
| | - Aydan İremnur Ergörün
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ali Doğan Dursun
- Department of Physiology, Faculty of Medicine, Atılım University, Ankara 06830, Turkey
| | - Zeynep Yiğman
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Neuroscience and Neurotechnology Center of Excellence, Gazi University, Ankara 06510, Turkey
| | - Metin Alkan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Life Sciences Application and Research Center, Gazi University, Ankara 06830, Turkey
- Laboratory Animal Breeding and Experimental Research Center, Gazi University, Ankara 06510, Turkey
| |
Collapse
|
4
|
Zheng Y, Gao Y, Zhu W, Bai XG, Qi J. Advances in molecular agents targeting toll-like receptor 4 signaling pathways for potential treatment of sepsis. Eur J Med Chem 2024; 268:116300. [PMID: 38452729 DOI: 10.1016/j.ejmech.2024.116300] [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: 10/19/2023] [Revised: 01/23/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by an infection. Toll-like receptor 4 (TLR4) is activated by endogenous molecules released by injured or necrotic tissues. Additionally, TLR4 is remarkably sensitive to infection of various bacteria and can rapidly stimulate host defense responses. The TLR4 signaling pathway plays an important role in sepsis by activating the inflammatory response. Accordingly, as part of efforts to improve the inflammatory response and survival rate of patients with sepsis, several drugs have been developed to regulate the inflammatory signaling pathways mediated by TLR4. Inhibition of TLR4 signal transduction can be directed toward either TLR4 directly or other proteins in the TLR4 signaling pathway. Here, we review the advances in the development of small-molecule agents and peptides targeting regulation of the TLR4 signaling pathway, which are characterized according to their structural characteristics as polyphenols, terpenoids, steroids, antibiotics, anthraquinones, inorganic compounds, and others. Therefore, regulating the expression of the TLR4 signaling pathway and modulating its effects has broad prospects as a target for the treatment of lung, liver, kidneys, and other important organs injury in sepsis.
Collapse
Affiliation(s)
- Yunyun Zheng
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China
| | - Yingying Gao
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China
| | - Weiru Zhu
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China
| | - Xian-Guang Bai
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China.
| | - Jinxu Qi
- Medicine College of Pingdingshan University, Pingdingshan, Henan, 467000, China.
| |
Collapse
|
5
|
Gao X, Wu Y. Perioperative acute kidney injury: The renoprotective effect and mechanism of dexmedetomidine. Biochem Biophys Res Commun 2024; 695:149402. [PMID: 38159412 DOI: 10.1016/j.bbrc.2023.149402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Dexmedetomidine (DEX) is a highly selective and potent α2-adrenoceptor (α2-AR) agonist that is widely used as a clinical anesthetic to induce anxiolytic, sedative, and analgesic effects. In recent years, a growing body of evidence has demonstrated that DEX protects against acute kidney injury (AKI) caused by sepsis, drugs, surgery, and ischemia-reperfusion (I/R) in organs or tissues, indicating its potential role in the prevention and treatment of AKI. In this review, we summarized the evidence of the renoprotective effects of DEX on different models of AKI and explored the mechanism. We found that the renoprotective effects of DEX mainly involved antisympathetic effects, reducing inflammatory reactions and oxidative stress, reducing apoptosis, increasing autophagy, reducing ferroptosis, protecting renal tubular epithelial cells (RTECs), and inhibiting renal fibrosis. Thus, the use of DEX is a promising strategy for the management and treatment of perioperative AKI. The aim of this review is to further clarify the renoprotective mechanism of DEX to provide a theoretical basis for its use in basic research in various AKI models, clinical management, and the treatment of perioperative AKI.
Collapse
Affiliation(s)
- Xiong Gao
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yaohua Wu
- Department of Anesthesiology, Huanggang Central Hospital, Huanggang, Hube, China.
| |
Collapse
|
6
|
Mercantepe F, Tumkaya L, Mercantepe T, Akyildiz K, Ciftel S, Yilmaz A. The Effects of Dexmedetomidine on Abdominal Aortic Occlusion-Induced Ovarian Injury via Oxidative Stress and Apoptosis. Cells Tissues Organs 2023; 212:554-566. [PMID: 37339613 DOI: 10.1159/000531613] [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: 03/28/2023] [Accepted: 06/14/2023] [Indexed: 06/22/2023] Open
Abstract
Ischemia/reperfusion (I/R) induced ovarian damage is caused by various diseases such as ovarian torsion, ovarian transplantation, cardiovascular surgery, sepsis, or intra-abdominal surgery. I/R-related oxidative damage can impair ovarian functions, from oocyte maturation to fertilization. This study investigated the effects of dexmedetomidine (DEX), which has been shown to exhibit antiapoptotic, anti-inflammatory, and antioxidant effects, on ovarian I/R injury. We designed four study groups: group 1 (n = 6): control group; group 2 (n = 6): only DEX group; group 3 (n = 6): I/R group; group 4 (n = 6): I/R + DEX group. Then, ovarian samples were taken and examined histologically and immunohistochemically, and tissue malondialdehyde (MDA) and glutathione (GSH) levels were measured. In the I/R group MDA levels, caspase-3, NF-κB/p65, 8-OHdG positivity, and follicular degeneration, edema, and inflammation were increased compared to the control group (p = 0.000). In addition, GSH levels were significantly decreased in the I/R group compared to the control group (p = 0.000). On the other hand, in the I/R + DEX treatment group MDA levels, caspase-3, NF-κB/p65, 8-OHdG positivity, follicular degeneration, edema, and inflammation findings were decreased than in the I/R group (p = 0.000, p = 0.005, p = 0.005, p = 0.001, p = 0.005, respectively). However, GSH levels increased significantly in the I/R + DEX treatment group compared to the I/R group (p = 0.000). DEX protects against ovarian I/R injury through antioxidation and by suppressing inflammation and apoptosis.
Collapse
Affiliation(s)
- Filiz Mercantepe
- Department of Endocrinology and Metabolism Diseases, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Kerimali Akyildiz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Serpil Ciftel
- Department of Endocrinology and Metabolism Diseases, Erzurum Regional Education and Research Hospital, Erzurum, Turkey
| | - Adnan Yilmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
7
|
Yamaguchi Y, Hosokawa S, Haraguchi G, Kajikawa Y, Sakurai M, Ishii T, Ando N, Morio T, Doi S, Furukawa T. The Anti-Inflammatory Effects and Clinical Potential of Dexmedetomidine in Pulmonary Arterial Hypertension. J Pharmacol Exp Ther 2023; 385:88-94. [PMID: 36849413 DOI: 10.1124/jpet.122.001399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 03/01/2023] Open
Abstract
A pathogenic aspect of pulmonary arterial hypertension (PAH) is the aberrant pulmonary arterial smooth muscle cell (PASMC) proliferation. PASMC proliferation is significantly affected by inflammation. A selective α-2 adrenergic receptor agonist called dexmedetomidine (DEX) modulates specific inflammatory reactions. We investigated the hypothesis that anti-inflammatory characteristics of DEX could lessen PAH that monocrotaline (MCT) causes in rats. In vivo, male Sprague-Dawley rats aged 6 weeks were subcutaneously injected with MCT at a dose of 60 mg/kg. Continuous infusions of DEX (2 µg/kg per hour) were started via osmotic pumps in one group (MCT plus DEX group) at day 14 following MCT injection but not in another group (MCT group). Right ventricular systolic pressure (RVSP), right ventricular end-diastolic pressure (RVEDP), and survival rate significantly improved in the MCT plus DEX group compared with the MCT group [RVSP, 34 mmHg ± 4 mmHg versus 70 mmHg ± 10 mmHg; RVEDP, 2.6 mmHg ± 0.1 mmHg versus 4.3 mmHg ± 0.6 mmHg; survival rate, 42% versus 0% at day 29 (P < 0.01)]. In the histologic study, the MCT plus DEX group showed fewer phosphorylated p65-positive PASMCs and less medial hypertrophy of the pulmonary arterioles. In vitro, DEX dose-dependently inhibited human PASMC proliferation. Furthermore, DEX decreased the expression of interleukin-6 mRNA in human PASMCs treated with fibroblast growth factor 2 (FGF2). These consequences suggest that DEX improves PAH by inhibiting PASMC proliferation through its anti-inflammatory properties. Additionally, DEX may exert anti-inflammatory effects via blocking FGF2-induced nuclear factor κ B activation. SIGNIFICANCE STATEMENT: Dexmedetomidine, a selective α-2 adrenergic receptor agonist utilized as a sedative in the clinical setting, improves pulmonary arterial hypertension (PAH) by inhibiting pulmonary arterial smooth muscle cell proliferation through its anti-inflammatory effect. Dexmedetomidine may be a new PAH therapeutic agent with vascular reverse remodeling effect.
Collapse
Affiliation(s)
- Yohei Yamaguchi
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Susumu Hosokawa
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Go Haraguchi
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Yusuke Kajikawa
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Makito Sakurai
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Taku Ishii
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Noboru Ando
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Tomohiro Morio
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Shozaburo Doi
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| | - Tetsushi Furukawa
- Departments of Pediatrics (Y.Y., S.H., M.S., T.I., T.M.) and Bio-Informational Pharmacology (T.F.), Tokyo Medical and Dental University, Tokyo, Japan; Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan (G.H.); Department of Emergency, Saitama Children's Medical Center, Saitama, Japan (Y.K.); Division of Pathology, Tokyo Medical and Dental University, Tokyo, Japan (N.A.); and Department of Pediatrics, National Hospital Organization Disaster Medical Center, Tokyo, Japan (S.D.)
| |
Collapse
|
8
|
Mercantepe F, Tumkaya L, Mercantepe T, Rakici S. Histopathological evaluation of the effects of dexmedetomidine against pituitary damage ınduced by X-ray irradiation. Biomarkers 2023; 28:168-176. [PMID: 36453587 DOI: 10.1080/1354750x.2022.2154385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background: The present study, aimed to investigate the potential negative effects of x-ray radiation and the effects of the α2-adrenergic receptor agonist dexmedetomidine on the pituitary gland.Methods: Twenty-four Sprague-Dawley rats were divided into three groups: Rats in Group 1 (control group). Group 2 (X-ray irradiation) and group 3 (X-ray irradiation + Dexmedetomidine) were given a total of 10 Gy external beam total body irradiation. Group 3 was given a single intraperitoneal dose of 200 µg/kg dexmedetomidine 30 minutes before RT.Results: In sections obtained from the x-ray irradiation group, we observed many necrotic in adenohypophysis and neurohypophysis. In addition, there were extensive oedematous areas and vascular congestions due to the necrotic cells in both the adenohypophysis and neurohypophysis. In contrast, we observed a reduction in necrotic chromophobic and chromophilic cells in adenohypophyseal tissue and a reduction in necrotic pituicytes in neurohypophyseal tissue in the dexmedetomidine treatment group. In addition, we determined lower caspase-3 and TUNEL expression in the dexmedetomidine treatment group compared with the x-ray irradiation group. Dexmedetomidine reduced x-ray radiation-induced pituitary damage by preventing apoptosis.Conclusions: The present study demonstrated the use of dexmedetomidine in situations related to radiation toxicity and offers the potential for a comprehensive study.
Collapse
Affiliation(s)
- Filiz Mercantepe
- Department of Endocrinology and Metabolism Diseases, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Sema Rakici
- Department of Radiation Oncology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
9
|
Han Z, Yuan Z, Shu L, Li T, Yang F, Chen L. Extracellular histone H3 facilitates ferroptosis in sepsis through ROS/JNK pathway. Immun Inflamm Dis 2023; 11:e754. [PMID: 36705411 PMCID: PMC9795329 DOI: 10.1002/iid3.754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/27/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Previous evidence realized the critical role of histone in disease control. The anti-inflammatory function of estradiol (E2) in sepsis has been documented. We here intended to unveil the role of extracellular histone H3 in sepsis regarding cell ferroptosis and the role of E2 in a such mechanism. METHODS Clinical sample, cecal ligation and puncture (CLP)-induced animal models and lipopolysaccharides (LPS)-induced cell models were prepared for testing relative expression of extracellular histone H3 and E2 as well as analyzing the role of extracellular histone H3 and E2 in sepsis concerning cell viability, reactive oxygen species (ROS), and ferroptosis. RESULTS Under sepsis, we found increased ferroptosis and extracellular histone H3 content, but reduced E2 concentration. Extracellular histone H3 facilitated ferroptosis of human umbilical vein endothelial cells (HUVECs) induced by LPS through activating the ROS/c-Jun N-terminal kinase (JNK) pathway. Moreover, E2 antagonized the effect of extracellular histone H3 on LPS-induced HUVEC ferroptosis and sepsis injury in CLP-induced animal models. CONCLUSION We highlighted that extracellular histone H3 facilitated lipopolysaccharides-induced HUVEC ferroptosis via activating ROS/JNK pathway, and such an effect could be antagonized by E2.
Collapse
Affiliation(s)
- Zhijun Han
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Zhizhou Yuan
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Linfei Shu
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Tao Li
- The Second Affiliated Hospital of Hainan Medical UniversityHaikouHainan ProvinceChina
| | - Fan Yang
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Lei Chen
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| |
Collapse
|
10
|
Carnicelli P, Otsuki DA, Monteiro Filho A, Kahvegian MAP, Ida KK, Auler-Jr JOC, Rouby JJ, Fantoni DT. Effects of dexmedetomidine on hemodynamic, oxygenation, microcirculation, and inflammatory markers in a porcine model of sepsis. Acta Cir Bras 2022; 37:e370703. [PMID: 36383809 DOI: 10.1590/acb370703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE To determine whether dexmedetomidine aggravates hemodynamic, metabolic variables, inflammatory markers, and microcirculation in experimental septic shock. METHODS Twenty-four pigs randomized into: Sham group (n = 8), received saline; Shock group (n = 8), received an intravenous infusion of Escherichia coli O55 (3 × 109 cells/mL, 0.75 mL/kg, 1 hour); Dex-Shock group (n = 8), received bacteria and intravenous dexmedetomidine (bolus 0.5 mcg/kg followed by 0.7 mcg/kg/h). Fluid therapy and/ornorepinephrine were administered to maintain a mean arterial pressure > 65 mmHg. Hemodynamic, metabolic, oxygenation, inflammatory markers, and microcirculation were assessed at baseline, at the end of bacterial infusion, and after 60, 120, 180, and 240 minutes. RESULTS Compared to Shock group, Dex-Shock group presented a significantly increased oxygen extraction ratio at T180 (23.1 ± 9.7 vs. 32.5 ± 9.2%, P = 0.0220), decreased central venous pressure at T120 (11.6 ± 1 vs. 9.61 ± 1.2 mmHg, P = 0.0214), mixed-venous oxygen saturation at T180 (72.9 ± 9.6 vs. 63.5 ± 9.2%, P = 0.026), and increased plasma lactate (3.7 ± 0.5 vs. 5.5 ± 1 mmol/L, P = 0.003). Despite the Dex-Shock group having a better sublingual vessel density at T240 (12.5 ± 0.4 vs. 14.4 ± 0.3 mL/m2; P = 0.0003), sublingual blood flow was not different from that in the Shock group (2.4 ± 0.2 vs. 2.4 ± 0.1 mL/kg, P = 0.4418). CONCLUSIONS Dexmedetomidine did not worsen the hemodynamic, metabolic, inflammatory, or sublingual blood flow disorders resulting from septic shock. Despite inducing a better sublingual vessel density, dexmedetomidine initially and transitorily increased the mismatch between oxygen supply and demand.
Collapse
Affiliation(s)
- Paulo Carnicelli
- MSc. Universidade de São Paulo - Faculdade de Medicina Veterinária e Zootecnia - Surgery Department - São Paulo (SP), Brazil
| | - Denise Aya Otsuki
- PhD. Universidade de São Paulo - Hospital das Clínicas da Faculdade de Medicina - LIM08-Laboratory of Anesthesiology - São Paulo (SP), Brazil
| | | | | | - Keila Kazue Ida
- PhD. Texas A&M University - College of Veterinary Medicine and Biomedical Sciences - Department of Small Animal Clinical Sciences - College Station (TX), United States of America
| | - José Otavio Costa Auler-Jr
- PhD. Universidade de São Paulo - Hospital das Clínicas da Faculdade de Medicina - Laboratory of Anesthesiology - São Paulo (SP), Brazil
| | - Jean-Jacques Rouby
- PhD. Assistance Publique Hôpitaux de Paris - La Pitié Salpêtrière Hospital - Multidisciplinary Intensive Care Unit - Medicine Sorbonne University - Department of Anaesthesiology and Critical Care - Paris, France
| | - Denise Tabacchi Fantoni
- PhD. Universidade de São Paulo - Faculdade de Medicina Veterinária e Zootecnia - Surgery Department - São Paulo (SP), Brazil
| |
Collapse
|
11
|
Yin H, Feng Y, Duan Y, Ma S, Guo Z, Wei Y. Hydrogen gas alleviates lipopolysaccharide-induced acute lung injury and inflammatory response in mice. J Inflamm (Lond) 2022; 19:16. [PMID: 36253774 PMCID: PMC9575233 DOI: 10.1186/s12950-022-00314-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Background Chronic inflammation and oxidant/antioxidant imbalance are two main pathological features associated with lipopolysaccharide (LPS)-induced acute lung injury (ALI). The following study investigated the protective role of hydrogen (H2), a gaseous molecule without known toxicity, in LPS-induced lung injury in mice and explored its potential molecular mechanisms. Methods Mice were randomly divided into three groups: H2 control group, LPS group, and LPS + H2 group. The mice were euthanized at the indicated time points, and the specimens were collected. The 72 h survival rates, cytokines contents, pathological changes, expression of Toll-like receptor 4 (TLR4), and oxidative stress indicators were analyzed. Moreover, under different culture conditions, RAW 264.7 mouse macrophages were used to investigate the potential molecular mechanisms of H2 in vitro. Cells were divided into the following groups: PBS group, LPS group, and LPS + H2 group. The cell viability, intracellular ROS, cytokines, and expression of TLR4 and nuclear factor kappa-B (NF-κB) were observed. Results Hydrogen inhalation increased the survival rate to 80%, reduced LPS-induced lung damage, and decreased inflammatory cytokine release in LPS mice. Besides, H2 showed remarked anti-oxidative activity to reduce the MDA and NO contents in the lung. In vitro data further indicated that H2 down-regulates the levels of ROS, NO, TNF-α, IL-6, and IL-1β in LPS-stimulated macrophages and inhibits the expression of TLR4 and the activation of nuclear factor kappa-B (NF-κB). Conclusion Hydrogen gas alleviates lipopolysaccharide-induced acute lung injury and inflammatory response most probably through the TLR4-NF-κB pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12950-022-00314-x.
Collapse
Affiliation(s)
- Hongling Yin
- grid.24516.340000000123704535Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Yajing Feng
- grid.24516.340000000123704535Department of Center ICU, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Yi Duan
- grid.24516.340000000123704535Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Shaolin Ma
- grid.24516.340000000123704535Department of Critical Care Medicine, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Zhongliang Guo
- grid.452753.20000 0004 1799 2798Department of Respiratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| | - Youzhen Wei
- grid.24516.340000000123704535Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120 China
| |
Collapse
|
12
|
Senousy SR, Ahmed ASF, Abdelhafeez DA, Khalifa MMA, Abourehab MAS, El-Daly M. Alpha-Chymotrypsin Protects Against Acute Lung, Kidney, and Liver Injuries and Increases Survival in CLP-Induced Sepsis in Rats Through Inhibition of TLR4/NF-κB Pathway. Drug Des Devel Ther 2022; 16:3023-3039. [PMID: 36105322 PMCID: PMC9467300 DOI: 10.2147/dddt.s370460] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Abstract Inflammation and oxidative stress play a major role in the development of sepsis and its associated complications, leading to multiple organ failure and death. The lungs, liver, and kidneys are among the early affected organs correlated with mortality in sepsis. Alpha-chymotrypsin (α-ch) is a serine protease that exerts anti-inflammatory, anti-edematous, and anti-oxidant properties. Purpose This study was undertaken to elucidate if the anti-inflammatory and anti-oxidant effects of α-ch observed in previous studies can alleviate lung, liver, and kidney injuries in a cecal ligation and puncture (CLP)-induced sepsis model, and thus decrease mortality. Materials and Methods Septic animals were given α-ch 2 h post CLP procedure. Sepsis outcomes were assessed in the lungs, liver, and kidneys. Separate animal groups were investigated for a survival study. Results CLP resulted in 0% survival, while α-chymotrypsin post-treatment led to 50% survival at the end of the study. Administration of α-chymotrypsin resulted in a significant attenuation of sepsis-induced elevated malonaldehyde (MDA) and total nitrite/nitrate (NOx) levels. In addition, there was a significant increase in reduced glutathione (GSH) content and superoxide dismutase (SOD) activity in the lungs, liver, and kidneys. Administration of α-ch reduced elevated tissue expression of toll-like receptor-4 (TLR4), nuclear factor kappa-B (NF-κB), myeloperoxidase (MPO), and inducible nitric oxide synthase (iNOS). Alpha-chymotrypsin resulted in a significant reduction in serum levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6). Alpha-chymotrypsin attenuated the rise in serum creatinine, cystatin C, blood urea nitrogen (BUN), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) levels that was observed in the septic group. In addition, α-ch significantly reduced the lung wet/dry weight ratio, total protein content, and leukocytic counts in bronchoalveolar lavage fluid (BALF). Histopathological examination of the lungs, liver, and kidneys confirmed the protective effects of α-ch on those organs. Conclusion α-ch has protective potential against sepsis through lowering tissue expression of TLR4, NF-κB, MPO, and iNOS leading to decreased oxidative stress and inflammatory signals induced by sepsis. This effect appeared to alleviate the damage to the lungs, liver, and kidneys and increase survival in rats subjected to sepsis.
Collapse
Affiliation(s)
- Shaymaa Ramzy Senousy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Correspondence: Al-Shaimaa F Ahmed, Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt, Tel +20 1020018842, Email
| | - Dalia A Abdelhafeez
- Department of Pathology, Faculty of Medicine, Minia University, Minia, Egypt
| | | | - Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| |
Collapse
|
13
|
Senousy SR, El-Daly M, Ibrahim ARN, Khalifa MMA, Ahmed ASF. Effect of Celecoxib and Infliximab against Multiple Organ Damage Induced by Sepsis in Rats: A Comparative Study. Biomedicines 2022; 10:biomedicines10071613. [PMID: 35884918 PMCID: PMC9312943 DOI: 10.3390/biomedicines10071613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 12/30/2022] Open
Abstract
In cases of sepsis, the immune system responds with an uncontrolled release of proinflammatory cytokines and reactive oxygen species. The lungs, kidneys, and liver are among the early impacted organs during sepsis and are a direct cause of mortality. The aim of this study was to compare the effects of infliximab (IFX) and celecoxib (CLX) on septic rats that went through a cecal ligation and puncture (CLP) surgery to induce sepsis. This study included four groups: sham, CLP (untreated), and CLP-treated with CLX or IFX. The administration of “low dose” CLX or IFX was performed after 2 h following the induction of sepsis. Twenty-four hours following the induction of sepsis, the rats were sacrificed and blood samples were collected to evaluate kidney, liver, and lung injuries. MDA and NOx content, in addition to SOD activity and GSH levels, were evaluated in the tissue homogenates of each group. Tissue samples were also investigated histopathologically. In a separate experiment, the same groups were employed to evaluate the survival of septic rats in a 7-day observation period. The results of this study showed that treatment with either CLX or IFX ameliorated the three organs’ damage compared to septic-untreated rats, decreased oxidative stress, enhanced the antioxidant defense, and reduced serum cytokines. As a result, a higher survival rate resulted: 62.5% and 37.5% after the administration of CLX and IFX, respectively, compared to 0% in the CLP group after 7 days. No significant differences were observed between the two agents in all measured parameters. Histopathological examination confirmed the observed results. In conclusion, CLX and IFX ameliorated lung, kidney, and liver injuries associated with sepsis through anti-inflammatory and antioxidant actions, which correlated to the increase in survival observed with both of them.
Collapse
Affiliation(s)
- Shaymaa Ramzy Senousy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61511, Egypt; (S.R.S.); (M.E.-D.); (M.M.A.K.); (A.-S.F.A.)
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61511, Egypt; (S.R.S.); (M.E.-D.); (M.M.A.K.); (A.-S.F.A.)
| | - Ahmed R. N. Ibrahim
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61511, Egypt
- Correspondence: ; Tel.: +96-65-5408-8979
| | - Mohamed Montaser A. Khalifa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61511, Egypt; (S.R.S.); (M.E.-D.); (M.M.A.K.); (A.-S.F.A.)
| | - Al-Shaimaa F. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61511, Egypt; (S.R.S.); (M.E.-D.); (M.M.A.K.); (A.-S.F.A.)
| |
Collapse
|
14
|
Gao F, Guo Z, Gao Y, Wang C, Wang H, Yao X, Shi B. Maternal oxidized soybean oil exposure in rats during lactation damages offspring kidneys via Nrf2/HO-1 and NF-κB signaling pathway. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3119-3129. [PMID: 34791653 DOI: 10.1002/jsfa.11653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 08/11/2021] [Accepted: 11/18/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Cooking oil is an indispensable component of the human diet. However, oils usually undergo thermal oxidation. Oxidized soybean oil (OSO) has been shown to have detrimental effects on humans and has emerged as a root cause of many chronic diseases. The objective of this work was to evaluate the effects of puerpera exposure to OSO on kidney damage in the mother and offspring using lactating rats as an experimental model. RESULTS Pathological sections and ultrastructure showed that OSO exposure resulted in various levels of damage to lactating rats and their offspring. OSO induced oxidative stress in the kidneys of lactating rats, as evidenced by increased levels of hydrogen peroxide, interleukin (IL)-1β, and IL-8. OSO increased the activities of glutathione peroxidase and superoxide dismutase. OSO upregulated the expression of apoptosis-related genes, nuclear factor-erythroid 2-related factor 2 (Nrf2), and nuclear factor κB-related inflammatory factor genes. In the offspring of the OSO-exposed mothers, hydrogen peroxide, malondialdehyde, IL-6, and tumor necrosis factor-alpha contents were increased. Furthermore, OSO enhanced the levels of Nrf2, NAD(P)H quinone oxidoreductase 1, heme oxygenase 1, and p65 and decreased B-cell lymphoma 2. CONCLUSION These findings indicated that the kidneys of two generations of rats were compromised by oxidative damage when fed OSO during lactation. This study provides evidence for increasing the genes expression of the Nrf2/heme oxygenase 1 pathway to alleviate the kidney damage caused by OSO in the mother and offspring. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Feng Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Zhiqiang Guo
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Yanan Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Chuanqi Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Huiting Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Xinxin Yao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
15
|
TANYELİ A, EKİNCİ AKDEMİR FN, ERASLAN E, GÜLER MC, ÖZBEK ŞEBİN S, GÜLÇİN İ. The possible useful effectiveness of sinapic acid in secondary organ damage sepsis-induced in rats. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2021. [DOI: 10.33808/clinexphealthsci.864015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
16
|
Ruegg G, Luethi N, Cioccari L. The Role of Dexmedetomidine for the Prevention of Acute Kidney Injury in Critical Care. EMJ NEPHROLOGY 2021:97-106. [DOI: 10.33590/emjnephrol/21-00087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
Acute kidney injury (AKI) occurs in up to 50% of patients admitted to the intensive care unit and is associated with increased mortality. Currently, there is no effective pharmacotherapy for prevention or treatment of AKI. In animal models of sepsis and ischaemia-reperfusion, α2-agonists like dexmedetomidine (DEX) exhibit anti-inflammatory properties and experimental data indicate a potential protective effect of DEX on renal function. However, clinical trials have yielded inconsistent results in critically ill patients. This review discusses the pathophysiological mechanisms involved in AKI, the renal effects of DEX in various intensive care unit-related conditions, and summarises the available literature addressing the use of DEX for the prevention of AKI.
Collapse
Affiliation(s)
- Gion Ruegg
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nora Luethi
- Department of Emergency Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Prahran, Australia
| | - Luca Cioccari
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Prahran, Australia
| |
Collapse
|
17
|
Li LC, Tian Y, Xiao J, Yang Y, Wu JN, Chen Y, Zhang PH, Gao-Smith F, Wang JG, Jin SW. Dexmedetomidine promotes inflammation resolving through TGF-β1 secreted by F4/80 +Ly6G + macrophage. Int Immunopharmacol 2021; 95:107480. [PMID: 33676148 DOI: 10.1016/j.intimp.2021.107480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/07/2023]
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist, which can regulate inflammatory responses. However, whether DEX interferes with the inflammation resolving remains unclear. Here, we reported the effects of DEX on zymosan-induced generalized inflammation in mice during resolution. Mice were administered intraperitoneally with DEX after the initiation of sepsis. The resolution interval (Ri), a vital resolution indice, decreased from twelve hours to eight hours after the administration of DEX. The induction of peritoneal pro-inflammatory interleukin [IL] - 1β and tumour necrosis factor-α (TNF-α) appeared to be inhibited. Of interest, the anti-inflammatory transforming growth factor-β1 (TGF-β1) but not IL-10 levels were up-regulated at twenty-four hours in the DEX group along with 1.0 mg/mice zymosan A (ZyA) treatment. The expression levels of multiple genes related to protective immune processes and clearance functions were detected and revealed the same trends. DEX markedly increased the F4/80+Ly6G+ macrophage population. Additionally, the adequate apoptotic neutrophil clearance from injury after DEX installation could be reverse by opsonization or co-instillation of TGF-β1 neutralizing antibody in vivo, promoting the inflammation-resolution programs. In conclusion, DEX post-treatment, via the increase of F4/80+Ly6G+ macrophages, provokes further secretion of TGF-β1, leading to the attenuated cytokine storm and accelerated inflammation resolving.
Collapse
Affiliation(s)
- Lin-Chao Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yang Tian
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Ji Xiao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yi Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Jin-Ni Wu
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China
| | - Yan Chen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Pu-Hong Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Fang Gao-Smith
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| | - Jian-Guang Wang
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China.
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| |
Collapse
|
18
|
Plummer MP, Lankadeva YR, Finnis ME, Harrois A, Harding C, Peiris RM, Okazaki N, May CN, Evans RG, Macisaac CM, Barge D, Bellomo R, Deane AM. Urinary and renal oxygenation during dexmedetomidine infusion in critically ill adults with mechanistic insights from an ovine model. J Crit Care 2021; 64:74-81. [PMID: 33794470 DOI: 10.1016/j.jcrc.2021.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Examine effects of dexmedetomidine on bladder urinary oxygen tension (PuO2) in critically ill patients and delineate mechanisms in an ovine model. MATERIALS AND METHODS In 12 critically ill patients: oxygen-sensing probe inserted in the bladder catheter and dexmedetomidine infusion at a mean (SD) rate of 0.9 ± 0.3 μg/kg/h for 24-h. In 9 sheep: implantation of flow probes around the renal and pulmonary arteries, and oxygen-sensing probes in the renal cortex, renal medulla and bladder catheter; dexmedetomidine infusion at 0.5 μg/kg/h for 4-h and 1.0 μg/kg/h for 4-h then 16 h observation. RESULTS In patients, dexmedetomidine decreased bladder PuO2at 2 (-Δ11 (95% CI 7-16)mmHg), 8 (-Δ 7 (0.1-13)mmHg) and 24 h (-Δ 11 (0.4-21)mmHg). In sheep, dexmedetomidine at 1 μg/kg/h reduced renal medullary oxygenation (-Δ 19 (14-24)mmHg) and bladder PuO2 (-Δ 12 (7-17)mmHg). There was moderate correlation between renal medullary oxygenation and bladder PuO2; intraclass correlation co-efficient 0.59 (0.34-0.80). Reductions in renal medullary oxygenation were associated with reductions in blood pressure, cardiac output and renal blood flow (P < 0.01). CONCLUSIONS Dexmedetomidine decreases PuO2in critically ill patients and in sheep. In sheep this reflects a decrease in renal medullary oxygenation, associated with reductions in cardiac output, blood pressure and renal blood flow.
Collapse
Affiliation(s)
- Mark P Plummer
- Department of Intensive Care, Royal Melbourne Hospital, 300 Grattan Street Parkville, Melbourne, Australia; Department of Critical Care, University of Melbourne, Melbourne, Australia.
| | - Yugeesh R Lankadeva
- Department of Critical Care, University of Melbourne, Melbourne, Australia; Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia.
| | - Mark E Finnis
- Department of Critical Care, University of Melbourne, Melbourne, Australia; Department of Intensive Care, Royal Adelaide Hospital, Adelaide, Australia.
| | - Anatole Harrois
- Department of Intensive Care, Royal Melbourne Hospital, 300 Grattan Street Parkville, Melbourne, Australia; Department of Anesthesia and Surgical Intensive Care, Paris-Saclay University, Bicêtre University Hospital, Le Kremlin Bicêtre, France
| | - Charlie Harding
- Department of Intensive Care, Royal Melbourne Hospital, 300 Grattan Street Parkville, Melbourne, Australia.
| | - Rachel M Peiris
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia.
| | - Nobuki Okazaki
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Clive N May
- Department of Critical Care, University of Melbourne, Melbourne, Australia; Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia.
| | - Roger G Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia.
| | - Christopher M Macisaac
- Department of Intensive Care, Royal Melbourne Hospital, 300 Grattan Street Parkville, Melbourne, Australia.
| | - Deborah Barge
- Department of Intensive Care, Royal Melbourne Hospital, 300 Grattan Street Parkville, Melbourne, Australia.
| | - Rinaldo Bellomo
- Department of Intensive Care, Royal Melbourne Hospital, 300 Grattan Street Parkville, Melbourne, Australia; Department of Critical Care, University of Melbourne, Melbourne, Australia.
| | - Adam M Deane
- Department of Intensive Care, Royal Melbourne Hospital, 300 Grattan Street Parkville, Melbourne, Australia; Department of Critical Care, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
19
|
Lankadeva YR, Shehabi Y, Deane AM, Plummer MP, Bellomo R, May CN. Emerging benefits and drawbacks of α 2 -adrenoceptor agonists in the management of sepsis and critical illness. Br J Pharmacol 2021; 178:1407-1425. [PMID: 33450087 DOI: 10.1111/bph.15363] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 12/29/2022] Open
Abstract
Agonists of α2 -adrenoceptors are increasingly being used for the provision of comfort, sedation and the management of delirium in critically ill patients, with and without sepsis. In this context, increased sympathetic and inflammatory activity are common pathophysiological features linked to multi-organ dysfunction, particularly in patients with sepsis or those undergoing cardiac surgery requiring cardiopulmonary bypass. Experimental and clinical studies support the notion that the α2 -adrenoceptor agonists, dexmedetomidine and clonidine, mitigate sympathetic and inflammatory overactivity in sepsis and cardiac surgery requiring cardiopulmonary bypass. These effects can protect vital organs, including the cardiovascular system, kidneys, heart and brain. We review the pharmacodynamic mechanisms by which α2 -adrenoceptor agonists might mitigate multi-organ dysfunction arising from pathophysiological conditions associated with excessive inflammatory and adrenergic stress in experimental studies. We also outline recent clinical trials that have examined the use of dexmedetomidine in critically ill patients with and without sepsis and in patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Yugeesh R Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.,Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Yahya Shehabi
- Department of Intensive Care Medicine, Monash Health, School of Clinical Sciences, Monash University, Melbourne, Prince of Wales Clinical School of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Adam M Deane
- Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia.,Department of Intensive Care Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Mark P Plummer
- Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia.,Department of Intensive Care Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Rinaldo Bellomo
- Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Clive N May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.,Centre for Integrated Critical Care, School of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Nephrology lacks effective therapeutics for many of the presentations and diseases seen in clinical practice. In recent decades, we have come to understand the central place of inflammation in initiating and propagating kidney disease, and, research in more recent years has established that the resolution of inflammation is a highly regulated and active process. With this, has evolved an appreciation that this aspect of the host inflammatory response is defective in kidney disease and led to consideration of a therapeutic paradigm aiming to harness the activity of the molecular drivers of the resolution phase of inflammation. Fatty-acid-derived Specialized pro-resolving mediators (SPMs), partly responsible for resolution of inflammation have gained traction as potential therapeutics. RECENT FINDINGS We describe our current understanding of SPMs for this purpose in acute and chronic kidney disease. These studies cement the place of inflammation and its defective resolution in the pathogenesis of kidney disease, and highlight new avenues for therapy. SUMMARY Targeting resolution of inflammation is a viable approach to treating kidney disease. We optimistically look forward to translating these experimental advances into tractable therapeutics to treat kidney disease.
Collapse
|
21
|
Anti-Inflammatory Activity of Diterpenoids from Celastrus orbiculatus in Lipopolysaccharide-Stimulated RAW264.7 Cells. J Immunol Res 2020; 2020:7207354. [PMID: 32802895 PMCID: PMC7414338 DOI: 10.1155/2020/7207354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
Celastrus orbiculatus Thunb has been known as an ethnopharmacological medicinal plant for antitumor, anti-inflammatory, and analgesic effects. Although various pharmacological studies of C. orbiculatus extract has been reported, an anti-inflammatory mechanism study of their phytochemical constituents has not been fully elucidated. In this study, compounds 1-17, including undescribed podocarpane-type trinorditerpenoid (3), were purified from C. orbiculatus and their chemical structure were determined by high-resolution electrospray ionization mass (HRESIMS) and nuclear magnetic resonance (NMR) spectroscopic data. To investigate the anti-inflammatory activity of compounds 1-17, nitric oxide (NO) secretion was evaluated in LPS-treated murine macrophages, RAW264.7 cells. Among compounds 1-17, deoxynimbidiol (1) and new trinorditerpenoid (3) showed the most potent inhibitory effects (IC50: 4.9 and 12.6 μM, respectively) on lipopolysaccharide- (LPS-) stimulated NO releases as well as proinflammatory mediators, such as inducible nitric oxide (iNOS), cyclooxygenase- (COX-) 2, interleukin- (IL-) 1β, IL-6, and tumor necrosis factor- (TNF-) α. Its inhibitory activity of proinflammatory mediators is contributed by suppressing the activation of nuclear transcription factor- (NF-) κB and mitogen-activated protein kinase (MAPK) signaling cascades including p65, inhibition of NF-κB (IκB), extracellular signal-regulated kinase (ERK), c-Jun NH2-terminal kinase (JNK), and p38. Therefore, these results demonstrated that diterpenoids 1 and 3 obtained from C. orbiculatus may be considered a potential candidate for the treatment of inflammatory diseases.
Collapse
|
22
|
Rehman A, Baloch NUA, Morrow JP, Pacher P, Haskó G. Targeting of G-protein coupled receptors in sepsis. Pharmacol Ther 2020; 211:107529. [PMID: 32197794 PMCID: PMC7388546 DOI: 10.1016/j.pharmthera.2020.107529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
The Third International Consensus Definitions (Sepsis-3) define sepsis as life-threatening multi-organ dysfunction caused by a dysregulated host response to infection. Sepsis can progress to septic shock-an even more lethal condition associated with profound circulatory, cellular and metabolic abnormalities. Septic shock remains a leading cause of death in intensive care units and carries a mortality of almost 25%. Despite significant advances in our understanding of the pathobiology of sepsis, therapeutic interventions have not translated into tangible differences in the overall outcome for patients. Clinical trials of antagonists of various pro-inflammatory mediators in sepsis have been largely unsuccessful in the past. Given the diverse physiologic roles played by G-protein coupled receptors (GPCR), modulation of GPCR signaling for the treatment of sepsis has also been explored. Traditional pharmacologic approaches have mainly focused on ligands targeting the extracellular domains of GPCR. However, novel techniques aimed at modulating GPCR intracellularly through aptamers, pepducins and intrabodies have opened a fresh avenue of therapeutic possibilities. In this review, we summarize the diverse roles played by various subfamilies of GPCR in the pathogenesis of sepsis and identify potential targets for pharmacotherapy through these novel approaches.
Collapse
Affiliation(s)
- Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Noor Ul-Ain Baloch
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - John P Morrow
- Department of Medicine, Columbia University, New York City, NY, United States
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York City, NY, United States.
| |
Collapse
|
23
|
Yang T, Feng X, Zhao Y, Zhang H, Cui H, Wei M, Yang H, Fan H. Dexmedetomidine Enhances Autophagy via α2-AR/AMPK/mTOR Pathway to Inhibit the Activation of NLRP3 Inflammasome and Subsequently Alleviates Lipopolysaccharide-Induced Acute Kidney Injury. Front Pharmacol 2020; 11:790. [PMID: 32670056 PMCID: PMC7326938 DOI: 10.3389/fphar.2020.00790] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
Background Acute kidney injury (AKI) is a severe complication of sepsis; however, no effective drugs have been found. Activation of the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome is a major pathogenic mechanism of AKI induced by lipopolysaccharide (LPS). Autophagy, a process of intracellular degradation related to renal homeostasis, effectively restricts inflammatory responses. Herein, we explored the potential protective mechanisms of dexmedetomidine (DEX), which has confirmed anti-inflammatory effects, on LPS-induced AKI. Methods AKI was induced in rats by injecting 10 mg/kg of LPS intraperitoneally (i.p.). Wistar rats received intraperitoneal injections of DEX (30 µg/kg) 30 min before an intraperitoneal injection of LPS. Atipamezole (ATI) (250 µg/kg) and 3-methyladenine (3-MA) (15 mg/kg) were intraperitoneally injected 30 min before the DEX injection. Results DEX significantly attenuated renal injury. Furthermore, DEX decreased activation of the NLRP3 inflammasome and expression of interleukins 1β and 18. In addition, autophagy-related protein and gene analysis indicated that DEX could significantly enhance autophagy. Finally, we verified the pharmacological effects of DEX on the 5′-adenosine monophosphate-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR) pathway. Atip and 3-MA significantly reversed the protective effects of DEX. Conclusions Our results suggest that the protective effects of DEX were mediated by enhanced autophagy via the α2-adrenoreceptor/AMPK/mTOR pathway, which decreased activation of the NLRP3 inflammasome. Above all, we verified the renal protective effects of DEX and offer a new treatment strategy for AKI.
Collapse
Affiliation(s)
- Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiujing Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haiyang Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hailin Cui
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mian Wei
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
24
|
Effect of Thymoquinone on Acute Kidney Injury Induced by Sepsis in BALB/c Mice. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1594726. [PMID: 32626733 PMCID: PMC7315249 DOI: 10.1155/2020/1594726] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/01/2020] [Indexed: 01/26/2023]
Abstract
Acute kidney injury (AKI) is a common complication of sepsis and has also been observed in some patients suffering from the new coronavirus pneumonia COVID-19, which is currently a major global concern. Thymoquinone (TQ) is one of the most active ingredients in Nigella sativa seeds. It has a variety of beneficial properties including anti-inflammatory and antioxidative activities. Here, we investigated the possible protective effects of TQ against kidney damage in septic BALB/c mice. Eight-week-old male BALB/c mice were divided into four groups: control, TQ, cecal ligation and puncture (CLP), and TQ+CLP. CLP was performed after 2 weeks of TQ gavage. After 48 h, we measured the histopathological alterations in the kidney tissue and the serum levels of creatinine (CRE) and blood urea nitrogen (BUN). We also evaluated pyroptosis (NLRP3, caspase-1), apoptosis (caspase-3, caspase-8), proinflammatory (TNF-α, IL-1β, and IL-6)-related protein and gene expression levels. Our results demonstrated that TQ inhibited CLP-induced increased serum CRE and BUN levels. It also significantly inhibited the high levels of NLRP3, caspase-1, caspase-3, caspase-8, TNF-α, IL-1β, and IL-6 induced by CLP. Furthermore, NF-κB protein level was significantly decreased in the TQ+CLP group than in the CLP group. Together, our results indicate that TQ may be a potential therapeutic agent for sepsis-induced AKI.
Collapse
|
25
|
Meng Q, Guo P, Jiang Z, Bo L, Bian J. Dexmedetomidine inhibits LPS-induced proinflammatory responses via suppressing HIF1α-dependent glycolysis in macrophages. Aging (Albany NY) 2020; 12:9534-9548. [PMID: 32433037 PMCID: PMC7288940 DOI: 10.18632/aging.103226] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
Dexmedetomidine, a highly selective α2-adrenoceptor agonist, has been reported to exert an anti-inflammatory effect in several animal models, but the mechanism remains unclear. Previous studies have shown that hypoxia inducible factor 1α-induced glycolysis is essential for the activation of inflammatory macrophages. However, whether dexmedetomidine influences hypoxia inducible factor 1α-induced glycolysis and thus exerts an anti-inflammatory effect has been poorly investigated. This study aims to elucidate the anti-inflammatory mechanism of dexmedetomidine involving the hypoxia inducible factor 1α-dependent glycolytic pathway. We showed that dexmedetomidine could suppress lipopolysaccharide-induced inflammatory cytokine production; inhibit the extracellular acidification rate, glucose consumption and lactate production; and decrease the expression of glycolytic genes in macrophages. The enhancement of glycolysis by the granulocyte-macrophage colony-stimulating factor or higher concentration of glucose could reverse the anti-inflammatory effect of dexmedetomidine on lipopolysaccharide-treated macrophages. Moreover, dexmedetomidine significantly inhibited the upregulation of hypoxia inducible factor 1α at the mRNA and protein levels. Genetic inhibition of hypoxia inducible factor 1α expression could reverse the anti-inflammatory effect of dexmedetomidine. Taken together, our results indicate that dexmedetomidine attenuates lipopolysaccharide-induced proinflammatory responses partially by suppressing hypoxia inducible factor 1α-dependent glycolysis in macrophages.
Collapse
Affiliation(s)
- Qingyuan Meng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Pinhao Guo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhengyu Jiang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
26
|
Wang C, Yuan W, Hu A, Lin J, Xia Z, Yang CF, Li Y, Zhang Z. Dexmedetomidine alleviated sepsis‑induced myocardial ferroptosis and septic heart injury. Mol Med Rep 2020; 22:175-184. [PMID: 32377745 PMCID: PMC7248514 DOI: 10.3892/mmr.2020.11114] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/17/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac dysfunction resulting from sepsis may cause significant morbidity and mortality, and ferroptosis plays a role in this pathology. Dexmedetomidine (Dex), a α2‑adrenergic receptor (α2‑AR) agonist exerts cardioprotective effects against septic heart dysfunction, but the exact mechanism is unknown. In the present study, sepsis was induced by cecal ligation and puncture (CLP) in male C57BL/6 mice. Dex and yohimbine hydrochloride (YOH), an α2‑AR inhibitor, were administered before inducing CLP. Then, 24 h after CLP, serum and heart tissue were collected to detect changes of troponin‑I (TN‑I), interleukin 6 (IL‑6), superoxide dismutase (SOD), malonaldehyde (MDA) and glutathione (GSH) levels, and iron release. Ferroptosis‑targeting proteins, apoptosis and inflammatory factors were assessed by western blotting or ELISA. It was found that, 24 h after CLP, TN‑I, a biomarker of myocardial injury, was significantly increased compared with the control group. Furthermore, the levels of MDA, 8‑hydroxy‑2'‑deoxyguanosine and the inflammatory factors IL‑6 and monocyte chemoattractant protein‑1 were also significantly increased. It was demonstrated that treatment with Dex reverted or attenuated these changes (CLP + Dex vs. CLP; P<0.05), but these protective effects of Dex were reversed by YOH. Moreover, CLP significantly decreased the protein expression levels of glutathione peroxidase 4 (GPX4), SOD and GSH. However, CLP increased expression levels of heme oxygenase‑1 (HO‑1), transferrin receptor, cleaved caspase 3, inducible nitric oxide synthase and gasdermin D, and iron concentrations. It was found that Dex reversed these changes, but YOH abrogated the protective effects of Dex (CLP + Dex + YOH vs. CLP + Dex; P<0.05). Therefore, the present results suggested that the attenuation of sepsis‑induced HO‑1 overexpression and iron concentration, and the reduction of ferroptosis via enhancing GPX4, may be the major mechanisms via which Dex alleviates sepsis‑induced myocardial cellular injury.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong 518000, P.R. China
| | - Wenlin Yuan
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong 518000, P.R. China
| | - Anmin Hu
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong 518000, P.R. China
| | - Juan Lin
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong 518000, P.R. China
| | - Zhengyuan Xia
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Catherine F Yang
- Department of Basic Sciences, College of Medicine, CA Northstate University, Elk Grove, CA 60009, USA
| | - Yalan Li
- Department of Anesthesiology, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510000, P.R. China
| | - Zhongjun Zhang
- Department of Anesthesiology, Shenzhen People's Hospital and Shenzhen Anesthesiology Engineering Center, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
27
|
Kiyonaga N, Moriyama T, Kanmura Y. Effects of dexmedetomidine on lipopolysaccharide-induced acute kidney injury in rats and mitochondrial function in cell culture. Biomed Pharmacother 2020; 125:109912. [DOI: 10.1016/j.biopha.2020.109912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/05/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022] Open
|
28
|
Human umbilical cord mesenchymal stem cell exosomes alleviate sepsis-associated acute kidney injury via regulating microRNA-146b expression. Biotechnol Lett 2020; 42:669-679. [PMID: 32048128 DOI: 10.1007/s10529-020-02831-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/03/2020] [Indexed: 12/29/2022]
Abstract
Human umbilical cord mesenchymal stem cell-derived exosomes (HucMSC-Ex) are a promising tool for the repair of acute kidney injury (AKI) caused by cisplatin and ischemia/reperfusion. However, the roles of hucMSC-Ex in sepsis-associated AKI repair and its mechanism are largely unknown. Hence, we constructed a sepsis model through cecal ligation and puncture (CLP), testing the benefits of hucMSC-Ex in the sepsis in terms of survival rate, serum renal markers levels, morphological changes and apoptosis. Immunohistochemistry staining and immunofluorescence assay were used to investigate the role of NF-κB activity in the repair of sepsis-associated AKI with hucMSC-Ex. HK-2 cells were transfected with microRNA-146b (miR-146b) mimics and inhibitors, respectively, and the regulatory effect of miR-146b on NF-κB activity was studied. We found that hucMSC-Ex treatment significantly decreased the serum creatinine (Cr) and blood urea nitrogen (BUN) levels, ameliorated the morphological damage and inhibited renal tubular cells apoptosis. More importantly, the survival rate at 72 h was 28% in CLP group and 45% in hucMSC-Ex group, respectively. Treatment with hucMSC-Ex improved survival in mice with sepsis. These effects of hucMSC-Ex were mediated by the inhibition of NF-κB activity and the lessening of pro-inflammatory response. Furthermore, hucMSC-Ex significantly increased miR-146b expression in kidney tissues. Conversely, interleukin (IL)-1 receptor-associated kinase (IRAK1) level, which is the target gene of miR-146b, clearly decreased in hucMSC-Ex group. In brief, this study showed that treatment with hucMSC-Ex decreased IRAK1 expression through the up-regulation of miR-146b level, led to the inhibition of NF-κB activity, and eventually alleviated sepsis-associated AKI and improved survival in mice with sepsis. HucMSC-Ex may be a novel therapeutic agent for the reduction of sepsis-associated AKI.
Collapse
|
29
|
Chai Y, Zhu K, Li C, Wang X, Shen J, Yong F, Jia H. Dexmedetomidine alleviates cisplatin‑induced acute kidney injury by attenuating endoplasmic reticulum stress‑induced apoptosis via the α2AR/PI3K/AKT pathway. Mol Med Rep 2020; 21:1597-1605. [PMID: 32016445 PMCID: PMC7003053 DOI: 10.3892/mmr.2020.10962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022] Open
Abstract
Cisplatin (CP) is an effective antineoplastic agent; however, CP-induced acute kidney injury (AKI) seriously affects the prognosis of patients with cancer. Endoplasmic reticulum (ER) stress (ERS)-induced apoptosis serves a pivotal role in the pathogenesis of CP-induced AKI. Dexmedetomidine (Dex), a potent α2 adrenergic agonist, has been reported to exert protective effects against AKI. However, the protective effects of Dex against CP-induced AKI and the potential molecular mechanisms remain unknown. In the present study, male Sprague-Dawley rats were divided into four groups (n=10/group), as follows: Control group; CP group, rats received an intraperitoneal (i.p.) injection of 5 mg/kg CP; Dex + CP group, rats received an i.p. injection of 25 µg/kg Dex immediately after CP treatment; and Dex + CP + atipamezole (Atip) group, rats received an i.p. injection of 250 µg/kg Atip, an α2 adrenoreceptor (α2AR) antagonist, and then received the same treatment as the Dex + CP group. Rats were anesthetized and sacrificed 96 h after CP injection. Subsequently, serum blood urea nitrogen (BUN) and serum creatinine (Scr) were analyzed, and kidney samples were collected for analyses. Pathological changes were examined using hematoxylin and eosin staining, and protein expression levels were assessed using western blotting and immunohistochemical staining. In addition, apoptosis was examined using a terminal deoxynucleotidyl transferase dUTP nick-end labeling assay. The present results suggested that Dex protected against CP-induced AKI by attenuating histological changes in the kidney, serum BUN and Scr production. Furthermore, the expression levels of 78-kDa glucose-regulated protein, C/EBP homologous protein and caspase-12, and the apoptotic rate in the kidney were decreased following Dex treatment. In addition, the expression levels of phosphorylated (p)-PI3K and p-AKT in the Dex + CP group were significantly increased. Conversely, the renoprotective effects of Dex were attenuated following the addition of Atip. In conclusion, Dex may alleviate CP-induced AKI by attenuating ERS-induced apoptosis, at least in part, via the α2AR/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yejing Chai
- Department of Medical Periodical Press, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Kangsheng Zhu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Chao Li
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaofan Wang
- Graduate School of Hebei Medical University, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Junmei Shen
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Fangfang Yong
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Huiqun Jia
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
30
|
Zeng M, Cao Y, Xu R, Wu Y, Wang Y, Zhang Y, Zheng X, Feng W. Oleanolic acid derivative isolated fromGardenia jasminoidesvar.radicansalleviates LPS-induced acute kidney injury in mice by reducing oxidative stress and inflammatory responsesviathe TLR4/NF-κB/NLRP3 signaling pathway. NEW J CHEM 2020. [DOI: 10.1039/c9nj05294a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Acute kidney injury (AKI) is a frequent complication of sepsis with hallmarks including inflammation and oxidative stress.
Collapse
Affiliation(s)
- Mengnan Zeng
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| | - Yangang Cao
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| | - Ruiqi Xu
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| | - Yuanyuan Wu
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| | - Yangyang Wang
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| | - Yanli Zhang
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| | - Xiaoke Zheng
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| | - Weisheng Feng
- Department of Pharmacy
- Henan University of Chinese Medicine
- Zhengzhou 450046
- China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province
| |
Collapse
|
31
|
Yao W, Guo A, Han X, Wu S, Chen C, Luo C, Li H, Li S, Hei Z. Aerosol inhalation of a hydrogen-rich solution restored septic renal function. Aging (Albany NY) 2019; 11:12097-12113. [PMID: 31841441 PMCID: PMC6949055 DOI: 10.18632/aging.102542] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/19/2019] [Indexed: 12/29/2022]
Abstract
Sepsis-related acute kidney injury (AKI) is known to be caused by inflammation. We explored the renal protective effects of aerosol inhalation of a hydrogen-rich solution (HRS; hydrogen gas dissolved to saturation in saline) in a mouse model of septic AKI. Septic AKI was induced through 18 hours of cecal ligation and puncture. AKI occurred during the early stage of sepsis, as evidenced by increased blood urea nitrogen and serum creatinine levels, pathological changes, renal fibrosis and renal tubular epithelial cell apoptosis, accompanied by macrophage infiltration and M1 macrophage-associated pro-inflammatory cytokine (Il-6 and Tnf-α) generation in renal tissues. Aerosol inhalation of the HRS increased anti-inflammatory cytokine (Il-4 and Il-13) mRNA levels in renal tissues and promoted macrophage polarization to the M2 type, which generated additional anti-inflammatory cytokines (Il-10 and Tgf-β). Ultimately, aerosol inhalation of HRS protected the kidneys and increased survival among septic mice. HRS was confirmed to promote M2 macrophage polarization in lipopolysaccharide-stimulated RAW 264.7 cells. The TGF-β1 receptor inhibitor SB-431542 partly reversed the effects of HRS on renal function, fibrosis, tubular epithelial cell apoptosis and senescence in mice. Thus, HRS aerosol inhalation appears highly useful for renal protection and inflammation reduction in septic AKI.
Collapse
Affiliation(s)
- Weifeng Yao
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Anshun Guo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.,Department of Anesthesiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Shan Wu
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Chaojin Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Chenfang Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Haobo Li
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China
| | - Shangrong Li
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
32
|
Qiu Z, Lu P, Wang K, Zhao X, Li Q, Wen J, Zhang H, Li R, Wei H, Lv Y, Zhang S, Zhang P. Dexmedetomidine Inhibits Neuroinflammation by Altering Microglial M1/M2 Polarization Through MAPK/ERK Pathway. Neurochem Res 2019; 45:345-353. [PMID: 31823113 DOI: 10.1007/s11064-019-02922-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 12/31/2022]
Abstract
Neuroinflammation is critical in the pathogenesis of neurological diseases. Microglial pro-inflammatory (M1) and anti-inflammatory (M2) status determines the outcome of neuroinflammation. Dexmedetomidine exerts anti-inflammatory effects in many neurological conditions. Whether dexmedetomidine functions via modulation of microglia M1/M2 polarization remains to be fully elucidated. In the present study, we investigated the anti-inflammatory effects of dexmedetomidine on the neuroinflammatory cell model and explored the potential mechanism. BV2 cells were stimulated with LPS to establish a neuroinflammatory model. The cell viability was determined with MTT assay. NO levels were assessed using a NO detection kit. The protein levels of IL-10, TNF-α, iNOS, CD206, ERK1/2, and pERK1/2 were quantified using Western blotting. LPS significantly increased pro-inflammatory factors TNF-α and NO, and M1 phenotypic marker iNOS, and decreased anti-inflammatory factor IL-10 and M2 phenotypic marker CD206 in BV2 cells. Furthermore, exposure of BV2 cells to LPS significantly raised pERK1/2 expression. Pretreatment with dexmedetomidine attenuated LPS-elicited changes in p-ERK, iNOS, TNF-α, NO, CD206 and IL-10 levels in BV2 cells. However, co-treatment with dexmedetomidine and LM22B-10, an agonist of ERK, reversed dexmedetomidine-elicited changes in p-ERK, iNOS, TNF-α, NO, CD206 and IL-10 levels in LPS-exposed BV2 cells. We, for the first time, showed that dexmedetomidine increases microglial M2 polarization by inhibiting phosphorylation of ERK1/2, by which it exerts anti-inflammatory effects in BV2 cells.
Collapse
Affiliation(s)
- Zhengguo Qiu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China.,Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Medical University, Shaanxi, 710038, China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Xijuan Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Qianqian Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Jieqiong Wen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Yuying Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Shuyue Zhang
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
33
|
Sun S, Wang J, Wang J, Wang F, Yao S, Xia H. Maresin 1 Mitigates Sepsis-Associated Acute Kidney Injury in Mice via Inhibition of the NF-κB/STAT3/MAPK Pathways. Front Pharmacol 2019; 10:1323. [PMID: 31787899 PMCID: PMC6855000 DOI: 10.3389/fphar.2019.01323] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/15/2019] [Indexed: 12/31/2022] Open
Abstract
Acute kidney injury (AKI) is one of the most common and serious complications of sepsis in which the inflammatory cascade plays a crucial role. There is now increasing evidence that lipid mediators derived from the omega-3 fatty acid docosahexaenoic acid (DHA) have potent anti-inflammatory effects that promote the timely regression of acute inflammation. In this study, we investigated the protective effects and molecular mechanism of a novel DHA-derived lipid mediator Maresin 1 (MaR1) on AKI in septic mice. The cecal ligation and puncture (CLP) was used to establish a sepsis mice model. As a result, we found that MaR1 significantly increased the 7-day survival rate of septic mice and the anti-inflammatory factor IL-10 while reducing bacterial load and pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β). In addition, MaR1 dose dependently reduced renal injury scores and serum creatinine and urea nitrogen levels in septic mice while inhibiting renal neutrophil infiltration and myeloperoxidase (MPO) activity. In terms of signaling pathway, we found that MaR1 inhibits the expression of phosphorylated p65, Stat3, JNK, ERK, and p38 and significantly reduces nuclear translocation of p65. In conclusion, our results indicate that MaR1 is able to reduce neutrophil infiltration and inhibit nuclear factor-kappa B/signal transducer and activator of transcriptor 3/mitogen-activated protein kinase (NF-κB/STAT3/MAPK) activity and regulate inflammatory cytokine level to inhibit inflammatory response and thereby weaken sepsis-associated AKI in mice.
Collapse
Affiliation(s)
- ShuJun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - JiaMei Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - JingXu Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - FuQuan Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - ShangLong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - HaiFa Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Dexmedetomidine reduces norepinephrine requirements and preserves renal oxygenation and function in ovine septic acute kidney injury. Kidney Int 2019; 96:1150-1161. [DOI: 10.1016/j.kint.2019.06.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/29/2022]
|
35
|
Zi SF, Li JH, Liu L, Deng C, Ao X, Chen DD, Wu SZ. Dexmedetomidine-mediated protection against septic liver injury depends on TLR4/MyD88/NF-κB signaling downregulation partly via cholinergic anti-inflammatory mechanisms. Int Immunopharmacol 2019; 76:105898. [PMID: 31520992 DOI: 10.1016/j.intimp.2019.105898] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Uncontrolled inflammatory responses exacerbate the pathogenesis of septic acute liver injury (ALI), posing a lethal threat to the host. Dexmedetomidine (DEX) has been reported to possess protective properties in inflammatory conditions. This study aimed to investigate whether DEX pretreatment exhibits hepatoprotection against ALI induced by lipopolysaccharide (LPS) in rats and determine its possible molecular mechanism. METHODS Septic ALI was induced by intravenous injection of LPS. The rats received DEX intraperitoneally 30 min before LPS administration. α-Bungarotoxin (α-BGT), a specific α7 nicotinic acetylcholine receptor (α7nAChR) antagonist, was administered intraperitoneally 1 h before LPS exposure. The role of the vagus nerve was verified by performing unilateral cervical vagotomy or sham surgery before sepsis. RESULTS The expression of α7nAChR, toll-like receptor 4 (TLR4), high mobility group box 1 (HMGB1), and cleaved caspase-3 increased, peaking 24 h during sepsis. DEX enhanced α7nAChR activation and reduced TLR4 expression upon challenge with LPS. DEX significantly prevented LPS-induced ALI, which was associated with increased survival, the mitigation of pathological changes, the attenuation of inflammatory cytokine expression and apoptosis, and the downregulation of TLR4/MyD88/NF-κB pathway. Moreover, the hepatoprotective effect of DEX was abolished by α-BGT. Further investigation established that vagotomy, compared to sham surgery, triggered more severe pathogenic manifestations and higher proinflammatory cytokine levels. The inhibitory effects of DEX were shown in sham-operated rats but not in vagotomized rats. CONCLUSIONS Our data highlight the pivotal function of α7nAChR and intact vagus nerves in protecting against LPS-induced ALI through inhibiting the TLR4/MyD88/NF-κB signaling pathway upon pretreatment with DEX.
Collapse
Affiliation(s)
- Shuang-Feng Zi
- Department of Critical Care Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| | - Jing-Hui Li
- Department of Critical Care Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China.
| | - Lei Liu
- Department of Critical Care Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| | - Chao Deng
- Department of Critical Care Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| | - Xue Ao
- Department of Critical Care Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| | - Dan-Dan Chen
- Department of Critical Care Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| | - Sheng-Zan Wu
- Department of Critical Care Medicine, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| |
Collapse
|
36
|
Yao Y, Hu X, Feng X, Zhao Y, Song M, Wang C, Fan H. Dexmedetomidine alleviates lipopolysaccharide-induced acute kidney injury by inhibiting the NLRP3 inflammasome activation via regulating the TLR4/NOX4/NF-κB pathway. J Cell Biochem 2019; 120:18509-18523. [PMID: 31243816 DOI: 10.1002/jcb.29173] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023]
Abstract
Dexmedetomidine (DEX) prevents kidney damage caused by sepsis, but the mechanism of this effect remains unclear. In this study, the protective molecular mechanism of DEX in lipopolysaccharide (LPS)-induced acute kidney injury was investigated and its potential pharmacological targets from the perspective of inhibiting oxidative stress damage and the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome activation. Intraperitoneal injection of DEX (30 μg/kg) significantly improved LPS (10 mg/kg) induced renal pathological damage and renal dysfunction. DEX also ameliorated oxidative stress damage by reducing the contents of reactive oxygen species, malondialdehyde and hydrogen peroxide, and increasing the level of glutathione, as well as the activity of superoxide dismutase and catalase. In addition, DEX prevented nuclear factor-kappa B (NF-κB) activation and I-kappa B (IκB) phosphorylation, as well as the expressions of NLRP3 inflammasome-associated protein and downstream IL-18 and IL-1β. The messengerRNA (mRNA) and protein expressions of toll-like receptor 4 (TLR4), NADPH oxidase-4 (NOX4), NF-κB, and NLRP3 were also significantly reduced by DEX. Their expressions were further evaluated by immunohistochemistry, yielding results were consistent with the results of mRNA and protein detection. Interestingly, the protective effects of DEX were reversed by atipamezole-an alpha 2 adrenal receptor (α2 AR) inhibitor, whereas idazoxan-an imidazoline receptor (IR) inhibitor failed to reverse this change. In conclusion, DEX attenuated LPS-induced AKI by inhibiting oxidative stress damage and NLRP3 inflammasome activation via regulating the TLR4/NOX4/NF-κB pathway, mainly acting on the α2 AR rather than IR.
Collapse
Affiliation(s)
- Yujie Yao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xueyuan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiujing Feng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Manyu Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chaoran Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Honggang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,College of Veterinary Medicine, Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin, China
| |
Collapse
|
37
|
Zhang H, Sha J, Feng X, Hu X, Chen Y, Li B, Fan H. Dexmedetomidine ameliorates LPS induced acute lung injury via GSK-3β/STAT3-NF-κB signaling pathway in rats. Int Immunopharmacol 2019; 74:105717. [PMID: 31254953 DOI: 10.1016/j.intimp.2019.105717] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 06/12/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022]
Abstract
Acute lung injury (ALI) is a serious complication of sepsis and an important cause of death in intensive care. Studies have shown that DEX can inhibit inflammation. However, the anti-inflammatory effect and protective mechanism of DEX in lipopolysaccharide (LPS) induced ALI are still unclear. ALI model was established by intraperitoneal injection of LPS (10 mg/kg) in Sprague-Dawley (SD) male rats. Firstly, at 4, 6, 8, 12 and 24 h after LPS treatment, lung injury including pathologic histology, lung edema, and inflammation were detected. The optimal time point for lung injury was determined to be 12 h, at which time DEX was added to further test. Furthermore, STAT3 inhibitor (NSC74859) and GSK-3β inhibitor (SB216763) were added to verify the role of STAT3, GSK-3β and NF-κB in ameliorated ALI. Our results show that DEX pretreatment significantly decreased lung Wet-to-Dry weight (W/D) ratio and MPO activity and ameliorated LPS induced lung histopathological alterations. In addition, we confirmed that DEX can increased the phosphorylation of STAT3 and GSK-3β, and inhibit the phosphorylation of nuclear factor-κB (NF-κB) p65 in the inflammatory response induced by LPS. What's more, NSC74859 inhibited the phosphorylation of STAT3 and reversed the protect effect of DEX on LPS. SB216763 inhibited the phosphorylation of NF-κB and reversed the damage effect of LPS and plays the same anti-inflammatory effect as DEX. In summary, our data demonstrated that DEX can ameliorate ALI induced by LPS through GSK-3β/STAT3-NF-κB pathway.
Collapse
Affiliation(s)
- Huayun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jichen Sha
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiujing Feng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xueyuan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yongping Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Bei Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Honggang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, China.
| |
Collapse
|
38
|
Dardalas I, Stamoula E, Rigopoulos P, Malliou F, Tsaousi G, Aidoni Z, Grosomanidis V, Milonas A, Papazisis G, Kouvelas D, Pourzitaki C. Dexmedetomidine effects in different experimental sepsis in vivo models. Eur J Pharmacol 2019; 856:172401. [PMID: 31108055 DOI: 10.1016/j.ejphar.2019.05.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/07/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
Abstract
Sepsis is a major cause of death and the most common cause of death among critically ill, non-ICU patients. Dexmedetomidine (DEX), an <alpha>2 adrenergic receptor agonist, presents sympatholytic action in certain parts of the brain with anxiolytic, sedative, and pain killing effects. Additionally, through the activation of <alpha>7 nicotinic acetylcholine receptor receptors, DEX reduces cytokine transcription and inhibits inflammation, rendering it beneficial during septic conditions. Moreover, there is a lot of interest in designing experimental sepsis models, where the administration of DEX is evaluated for its impact on multiple systems. This review focuses on experimental studies published between 1999 to March 2019 that were using DEX administration in sepsis in vivo models. From these, 36 articles were selected and summarized. Overall results show evidence that DEX may decrease mortality and inhibit inflammation, as it enhances the activity of the immune system while reducing its systemic reaction and lowering cytokine concentrations. Moreover DEX succeeds to alleviate heart injury during sepsis, acting beneficially for microcirculation and shows a neuroprotective role by inhibiting apoptotic pathways. In addition, DEX appears to have a protective role for liver and spleen as well as a beneficial role for the function of lungs and kidneys as it reduces sepsis-induced injuries and apoptosis in intra-abdominal experimental sepsis models.
Collapse
Affiliation(s)
- Ioannis Dardalas
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eleni Stamoula
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Panagiotis Rigopoulos
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Faye Malliou
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Georgia Tsaousi
- Clinic of Anesthesiology and Intensive Care, AHEPA University Hospital, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54006, Thessaloniki, Greece
| | - Zoi Aidoni
- Clinic of Anesthesiology and Intensive Care, AHEPA University Hospital, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54006, Thessaloniki, Greece
| | - Vasileios Grosomanidis
- Clinic of Anesthesiology and Intensive Care, AHEPA University Hospital, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54006, Thessaloniki, Greece
| | - Antonios Milonas
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Georgios Papazisis
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Dimitrios Kouvelas
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Chryssa Pourzitaki
- Department of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| |
Collapse
|
39
|
Zhang WQ, Xu P, Zhan XH, Zheng P, Yang W. Efficacy of dexmedetomidine for treatment of patients with sepsis: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2019; 98:e15469. [PMID: 31045827 PMCID: PMC6504533 DOI: 10.1097/md.0000000000015469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND This meta-analysis aimed to evaluate the effect of dexmedetomidine on prognosis in patients with sepsis. METHODS Computer-related electronic databases were searched, including PubMed, Embase, Web of Science, the Cochrane Library, and the China National Knowledge Infrastructure, from the date of database construction to January 2019. Stata 12.0 was used to perform a meta-analysis of short-term mortality [intensive care unit (ICU) mortality or 28-day mortality], ICU length of stay, and mechanical ventilation. Mortality was expressed using risk ratio (RR) and 95% confidence interval (CI). ICU length of stay and mechanical ventilation were expressed as weighted mean difference (WMD) and 95% CIs. RESULTS We finally included 8 randomized controlled trials in this meta-analysis. Compared with the control group, the dexmedetomidine group had a lower occurrence of 28-day mortality (RR, 0.49; 95% CI, 0.35 to 0.69; P = .000) and ICU mortality (RR, 0.44; 95% CI, 0.23 to 0.84; P = .013). However, there was no statistically significant difference for the length of hospital stay (WMD, -0.05; 95% CI, -0.59 to 0.48; P = .840) and mechanical ventilation time (WMD, 1.05; 95% CI, -0.27 to 2.37; P = .392) between dexmedetomidine group and control group. CONCLUSIONS In patients with sepsis, dexmedetomidine can reduce the short-term mortality of patients, but could not shorten the ICU length of stay and mechanical ventilation time. More clinical randomized controlled trials are needed to verify the efficacy and safety of dexmedetomidine on the length of hospital stay and mechanical ventilation time.
Collapse
Affiliation(s)
- Wen-Qing Zhang
- Department of Intensive Care Unit, Jingjiang People's Hospital
| | - Po Xu
- Department of Intensive Care Unit, JingJiang Chinese Medicine Hospital, Jingjiang, Jiangsu Province, China
| | - Xiao-Hong Zhan
- Department of Intensive Care Unit, Jingjiang People's Hospital
| | - Peng Zheng
- Department of Intensive Care Unit, Jingjiang People's Hospital
| | - Wei Yang
- Department of Intensive Care Unit, Jingjiang People's Hospital
| |
Collapse
|
40
|
Feng X, Guan W, Zhao Y, Wang C, Song M, Yao Y, Yang T, Fan H. Dexmedetomidine ameliorates lipopolysaccharide-induced acute kidney injury in rats by inhibiting inflammation and oxidative stress via the GSK-3β/Nrf2 signaling pathway. J Cell Physiol 2019; 234:18994-19009. [PMID: 30919976 DOI: 10.1002/jcp.28539] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/19/2022]
Abstract
Acute kidney injury (AKI) is a frequent and serious complication of sepsis; however, there are currently no effective therapies. Inflammation and oxidative stress are the major mechanisms implicated in lipopolysaccharide (LPS)-induced AKI. Dexmedetomidine (DEX) has been reported to have remarkable anti-inflammatory and antioxidant effects. Here, we examined the renoprotective effects of DEX and potential underlying mechanisms in rats with LPS-induced AKI. We analyzed renal function and structure; serum inflammatory cytokine; renal oxidant and antioxidant levels; and renal expression of glycogen synthase kinase-3β (GSK-3β)/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway-related proteins in rats 4 hr after administration of LPS. Pretreatment with DEX improved renal function and significantly reduced the levels of inflammatory cytokines and oxidative stress markers. Treatment with DEX and the GSK-3β inhibitor SB216367 promoted phosphorylation of GSK-3β, induced Nrf2 nuclear translocation, and increased transcription of the Nrf2 target genes heme oxygenase-1 and NAD(P)H quinone oxidoreductase-1, primarily in renal tubules. Alpha-2-adrenergic receptor (α2-AR) antagonist atipamezole and imidazoline I 2 receptor (I 2 R) antagonist idazoxan reversed the effects of DEX. These results suggest that the renoprotective effects of DEX are mediated via α2-AR and I 2 R-dependent pathways that reduce inflammation and oxidative stress through GSK-3β/Nrf2 signaling.
Collapse
Affiliation(s)
- Xiujing Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Wei Guan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Chaoran Wang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Manyu Song
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yujie Yao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
41
|
Cx43 Inhibition Attenuates Sepsis-Induced Intestinal Injury via Downregulating ROS Transfer and the Activation of the JNK1/Sirt1/FoxO3a Signaling Pathway. Mediators Inflamm 2019; 2019:7854389. [PMID: 30948926 PMCID: PMC6425293 DOI: 10.1155/2019/7854389] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
Intestinal injury has long been considered to play a crucial role in the pathophysiology of sepsis and has even been characterized as the “motor” of it. Thus, we explored the effects of connexin43 (Cx43) on sepsis-induced intestinal injury in order to provide potential therapeutic strategies. Rat cecal ligation and puncture (CLP) models in vivo and cell models (IEC-6 cells) pretreated with LPS in vitro were used in the current study. Firstly, different methods, such as Cx43 inhibitors (18-α-GA and oleamide) or siRNA targeting Cx43 and N-acetyl cysteine (NAC) (a kind of ROS scavenger), were used to observe the effects of Cx43 channels mediating ROS transfer on intestinal injury. Secondly, the influence of ROS content on the activity of the JNK1/Sirt1/FoxO3a signaling pathway was explored through the application of NAC, sp600125 (a JNK1 inhibitor), and nicotinamide (a Sirt1 inhibitor). Finally, luciferase assays and ChIP were used to determine the direct regulation of FoxO3a on proapoptotic proteins, Bim and Puma. The results showed that sepsis-induced intestinal injury presented a dynamic change, coincident with the alternation of Cx43 expression. The inhibition of Cx43 attenuated CLP-induced intestinal injury in vivo and LPS-induced IEC-6 injury in vitro. The changes of Cx43 channel function regulated ROS transfer between the neighboring cells, which mediated the activation of the JNK1/Sirt1/FoxO3a signaling pathway. FoxO3a directly affected its downstream target genes, Bim and Puma, which are responsible for cell or tissue apoptosis. In summary, our results suggest that Cx43 inhibition suppresses ROS transfer and inactivates the JNK1/Sirt1/FoxO3a signaling pathway to protect against sepsis-induced intestinal injury.
Collapse
|
42
|
Dexmedetomidine protects against lipopolysaccharide-induced early acute kidney injury by inhibiting the iNOS/NO signaling pathway in rats. Nitric Oxide 2019; 85:1-9. [PMID: 30659917 DOI: 10.1016/j.niox.2019.01.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 12/14/2022]
Abstract
Increasing evidence has demonstrated that dexmedetomidine (DEX) possesses multiple pharmacological actions. Herein, we explored the protective effect and potential molecular mechanism of DEX on lipopolysaccharide (LPS)-induced early acute kidney injury (AKI) from the perspective of antioxidant stress. We found that DEX (30 μg/kg, i.p.) ameliorated the renal dysfunction and histopathological damage (tubular necrosis, vacuolar degeneration, infiltration of inflammatory cells and cast formation) induced by LPS (10 mg/kg). DEX also attenuated renal oxidative stress remarkably in LPS-induced early AKI, as evidenced by reduction in production of reactive nitrogen species, decreasing malondialdehyde levels, as well as increasing superoxide dismutase activity and glutathione content. DEX prevented activator protein-1 translocation, inhibited phosphorylation of I-kappa B (IκB) and activation of nuclear factor kappa B (NF-κB) in LPS-induced early AKI, as assessed by real-time quantitative polymerase chain reaction and protein levels of c-Jun, c-Fos, IκB and NF-κB. Notably, DEX pretreatment had the same effect as intraperitoneal injection of an inhibitor of inducible nitric oxide synthase inhibitor (1400W; 15 mg/kg), and inhibited the activity of renal inducible nitric oxide synthase (iNOS) and decreased the expression of iNOS mRNA and NO production. However, the protective effect of DEX on LPS-induced early AKI was reversed by the alpha 2 adrenal receptor (α2-AR) inhibitor atipamezole, whereas the imidazoline receptor inhibitor idazoxan did not. Taken together, DEX protects against LPS-induced early AKI in rats by inhibiting the iNOS/NO signaling pathway, mainly by acting on α2-ARs instead of IRs.
Collapse
|