1
|
Pereira-Silva GC, Cornélio CKCA, Pacheco G, Rochael NC, Gomes IAB, Cajado AG, Silva KC, Gonçalves BS, Temerozo JR, Bastos RS, Rocha JA, Souza LP, Souza MHLP, Lima-Júnior RCP, Medeiros JVR, Filgueiras MC, Bou-Habib DC, Saraiva EM, Nicolau LAD. Diminazene aceturate inhibits the SARS-CoV-2 spike protein-induced inflammation involving leukocyte migration and DNA extracellular traps formation. Life Sci 2024; 352:122895. [PMID: 38986896 DOI: 10.1016/j.lfs.2024.122895] [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: 04/12/2024] [Revised: 06/16/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
AIMS To investigate the SARS-CoV-2 Spike protein (Spk)-induced inflammatory response and its downmodulation by diminazene aceturate (DIZE). MATERIALS AND METHODS Through inducing Spk inflammation in murine models, leukocyte migration to the peritoneum, levels of myeloperoxidase (MPO), malondialdehyde (MDA), rolling and adhesion of mesenteric leukocytes, and vascular permeability were investigated. Extracellular DNA traps (DETs) induced by Spk and the production of IL-6 and TNF-α were analyzed using human neutrophils, monocytes, and macrophages. In silico assays assessed the molecular interaction between DIZE and molecules related to leukocyte migration and DETs induction. KEY FINDINGS Spk triggered acute inflammation, demonstrated by increasing leukocyte migration. Oxidative stress was evidenced by elevated levels of MPO and MDA in the peritoneal liquid. DIZE attenuated cell migration, rolling, and leukocyte adhesion, improved vascular barrier function, mitigated DETs, and reduced the production of Spk-induced pro-inflammatory cytokines. Computational studies supported our findings, showing the molecular interaction of DIZE with targets such as β2 integrin, PI3K, and PAD2 due to its intermolecular coupling. SIGNIFICANCE Our results outline a novel role of DIZE as a potential therapeutic agent for mitigating Spk-induced inflammation.
Collapse
Affiliation(s)
- Gean C Pereira-Silva
- Laboratory on Innate Immunity, Department of Immunology, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Cassia K C A Cornélio
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil
| | - Gabriella Pacheco
- Department of Biochemistry and Pharmacology, Health Sciences Center, Universidade Federal do Piauí (UFPI), Teresina, PI, Brazil
| | - Natalia C Rochael
- Laboratory on Innate Immunity, Department of Immunology, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Isaac A B Gomes
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil
| | - Aurilene G Cajado
- Department of Physiology and Pharmacology, Universidade Federal do Ceará (UFC), Fortaleza, CE, Brazil
| | - Katriane C Silva
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil
| | | | - Jairo R Temerozo
- Laboratory on Thymus Research, Oswaldo Cruz Institute (Fiocruz), Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology on Neuroimmunemodulation, Rio de Janeiro, Brazil
| | - Ruan S Bastos
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil
| | - Jefferson A Rocha
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil
| | - Leonardo P Souza
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil
| | - Marcellus H L P Souza
- Department of Physiology and Pharmacology, Universidade Federal do Ceará (UFC), Fortaleza, CE, Brazil
| | - Roberto C P Lima-Júnior
- Department of Physiology and Pharmacology, Universidade Federal do Ceará (UFC), Fortaleza, CE, Brazil
| | - Jand V R Medeiros
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil; Department of Biochemistry and Pharmacology, Health Sciences Center, Universidade Federal do Piauí (UFPI), Teresina, PI, Brazil
| | - Marcelo C Filgueiras
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute (Fiocruz), Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology on Neuroimmunemodulation, Rio de Janeiro, Brazil
| | - Elvira M Saraiva
- Laboratory on Innate Immunity, Department of Immunology, Institute of Microbiology Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
| | - Lucas A D Nicolau
- Biotechnology and Biodiversity Center Research, Laboratory of Inflammation and Translational Gastroenterology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba, PI, Brazil; Department of Biochemistry and Pharmacology, Health Sciences Center, Universidade Federal do Piauí (UFPI), Teresina, PI, Brazil.
| |
Collapse
|
2
|
Suleimani YA, Maskari RA, Ali BH, Ali H, Manoj P, Al-Khamiyasi A, Abdelrahman AM. Nephroprotective effects of diminazene on doxorubicin-induced acute kidney injury in rats. Toxicol Rep 2023; 11:460-468. [PMID: 38053572 PMCID: PMC10693989 DOI: 10.1016/j.toxrep.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023] Open
Abstract
This study aimed to investigate the potential protective effects of diminazene, an activator of angiotensin II converting enzyme (ACE2), on kidney function and structure in rats with acute kidney injury (AKI) induced by the anticancer drug doxorubicin (DOX). The impact of diminazene was compared to that of two other drugs: the ACE inhibitor lisinopril and the angiotensin II type 1 (AT1) receptor blocker valsartan. Rats were subjected to a single intraperitoneal injection of DOX (13.5 mg/kg) on the 5th day, either alone or in combination with diminazene (15 mg/kg/day), lisinopril (10 mg/kg/day), or valsartan (30 mg/kg/day) for 8 consecutive days. Various markers related to kidney function, oxidative stress, and inflammation were measured in plasma and urine. Additionally, kidney tissues were assessed histopathologically. DOX-induced nephrotoxicity was confirmed by elevated levels of plasma urea, creatinine, and neutrophil gelatinase-associated lipocalin (NGAL). DOX also led to increased urinary N-acetyl-β-D-glucosaminidase (NAG) activity and decreased creatinine clearance, albumin levels, and osmolality. Moreover, DOX caused a reduction in renal oxidative stress markers, including superoxide dismutase (SOD), glutathione reductase (GR), and catalase activities, while increasing malondialdehyde (MDA) levels. It also raised plasma inflammatory markers, tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β). Concurrently administering diminazene significantly mitigated these DOX-induced changes, including histopathological alterations like renal tubule necrosis, tubular casts, shrunken glomeruli, and increased renal fibrosis. Similar protective effects were observed with lisinopril and valsartan. These protective effects, at least in part, appear to result from the anti-inflammatory and antioxidant properties of these drugs. In summary, this study suggests that the administration of diminazene, lisinopril, or valsartan had comparable effects in ameliorating the biochemical and histopathological aspects of DOX-induced acute kidney injury in rats.
Collapse
Affiliation(s)
- Yousuf Al Suleimani
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Raya Al Maskari
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Badreldin H. Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Haytham Ali
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Priyadarsini Manoj
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Ali Al-Khamiyasi
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| | - Aly M. Abdelrahman
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P.O. Box 35, Al Khod 123, Oman
| |
Collapse
|
3
|
Chen F, Wang Y, Chen Y, Fan J, Zhang C, He X, Yang X. JNK molecule is a toxic target for IPEC-J2 cell barrier damage induced by T-2 toxin. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115247. [PMID: 37453270 DOI: 10.1016/j.ecoenv.2023.115247] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
The most prevalent contaminated mycotoxin in feed and grain is T-2 toxin. The T-2 toxin's primary action target is the gut because it is the main organ of absorption. T-2 toxin can cause intestinal damage, but, few molecular mechanisms have been elucidated. It is important to discover the key pathways by which T-2 toxin causes enterotoxicity. In this research, IPEC-J2 cells are used as a cell model to investigate the function of the MAPK signaling pathway in T-2 toxin-induced intestinal epithelial cell damage. Throughout this research, T-2 toxin results in functional impairment in IPEC-J2 cells by reducing the TJ proteins Claudin, Occludin-1, ZO-1, N-cadherin, and CX-43 expression. T-2 toxin significantly reduced the survival of IPEC-J2 cells and increased LDH release in a dose-dependent way. T-2 toxin induced IPEC-J2 cell oxidative stress by raising ROS and MDA content, and mitochondrial damage was indicated by a decline in MMP and an increase in the opening degree of MPTP. T-2 toxin upregulated the expression of ERK, P38 and JNK, which triggered the MAPK signaling pathway. In addition, T-2 toxin caused IPEC-J2 cell inflammation responses reflected by increased the levels of inflammation-related factors IL-8, p65, P-p65 and IL-6, and down-regulated IL-10 expression level. Inhibition JNK molecule can ease IPEC-J2 cell functional impairment and inflammatory response. In conclusion, as a consequence of the T-2 toxin activating the JNK molecule, oxidative stress and mitochondrial damage are induced, which impair cellular inflammation.
Collapse
Affiliation(s)
- Fengjuan Chen
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002 Henan, China
| | - Youshuang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002 Henan, China
| | - Yunhe Chen
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002 Henan, China
| | - Jiayan Fan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002 Henan, China
| | - Cong Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002 Henan, China
| | - Xiuyuan He
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002 Henan, China
| | - Xu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002 Henan, China.
| |
Collapse
|
4
|
Zhang K, Li C, Sun J, Tian X. PRMT5 inhibition ameliorates inflammation and promotes the osteogenic differentiation of LPS‑induced periodontal stem cells via STAT3/NF‑κB signaling. Exp Ther Med 2023; 25:264. [PMID: 37206565 PMCID: PMC10189754 DOI: 10.3892/etm.2023.11963] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/06/2022] [Indexed: 05/21/2023] Open
Abstract
It has been reported that protein arginine methyltransferase 5 (PRMT5) serves a significant role in osteogenic differentiation and inflammatory response. Nevertheless, its role in periodontitis as well as its underlying mechanism remain to be elucidated. The aim of the present study was to explore the role of PRMT5 in periodontitis and whether PRMT5 could reduce liposaccharide (LPS)-induced inflammation of human periodontal ligament stem cells (hPDLSCs) and promote osteogenic differentiation through STAT3/NF-κB signaling. In the current study, the expression levels of PRMT5 were determined in LPS-induced hPDLSCs by reverse transcription-quantitative PCR and western blot analysis. ELISA and western blot analysis were employed to assess the secretion and expression levels of inflammatory factors, respectively. The osteogenic differentiation and mineralization potential of hPDLSCs were evaluated using alkaline phosphatase (ALP) activity assay, Alizarin red staining and western blot analysis. Additionally, western blot analysis was applied to determine the expression levels of the STAT3/NF-κB signaling pathway-related proteins. The results showed that the expression levels of PRMT5 were significantly enhanced in LPS-induced hPDLSCs. Additionally, PRMT5 knockdown reduced the contents of IL-1β, IL-6, TNF-α, inducible nitric oxide synthase and cyclooxygenase-2. PRMT5 depletion also enhanced ALP activity, improved the mineralization ability and upregulated bone morphogenetic protein 2, osteocalcin and runt-related transcription factor 2 in LPS-induced hPDLSCs. Furthermore, PRMT5 knockdown inhibited inflammation and promoted the osteogenic differentiation of hPDLSCs via blocking the activation of the STAT3/NF-κB signaling pathway. In conclusion, PRMT5 inhibition suppressed LPS-induced inflammation and accelerated osteogenic differentiation in hPDLSCs via regulating STAT3/NF-κB signaling, thus providing a potential targeted therapy for the improvement of periodontitis.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Changshun Li
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jian Sun
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Xiaobei Tian
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- Correspondence to: Professor Xiaobei Tian, Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, 130 Huaihai West Road, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
5
|
Hulme J. COVID-19 and Diarylamidines: The Parasitic Connection. Int J Mol Sci 2023; 24:6583. [PMID: 37047556 PMCID: PMC10094973 DOI: 10.3390/ijms24076583] [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: 03/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
As emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants (Omicron) continue to outpace and negate combinatorial vaccines and monoclonal antibody therapies targeting the spike protein (S) receptor binding domain (RBD), the appetite for developing similar COVID-19 treatments has significantly diminished, with the attention of the scientific community switching to long COVID treatments. However, treatments that reduce the risk of "post-COVID-19 syndrome" and associated sequelae remain in their infancy, particularly as no established criteria for diagnosis currently exist. Thus, alternative therapies that reduce infection and prevent the broad range of symptoms associated with 'post-COVID-19 syndrome' require investigation. This review begins with an overview of the parasitic-diarylamidine connection, followed by the renin-angiotensin system (RAS) and associated angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSSR2) involved in SARS-CoV-2 infection. Subsequently, the ability of diarylamidines to inhibit S-protein binding and various membrane serine proteases associated with SARS-CoV-2 and parasitic infections are discussed. Finally, the roles of diarylamidines (primarily DIZE) in vaccine efficacy, epigenetics, and the potential amelioration of long COVID sequelae are highlighted.
Collapse
Affiliation(s)
- John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
6
|
Kulhari U, Kundu S, Mugale MN, Sahu BD. Nuciferine alleviates intestinal inflammation by inhibiting MAPK/NF-κB and NLRP3/Caspase 1 pathways in vivo and in vitro. Int Immunopharmacol 2023; 115:109613. [PMID: 36577154 DOI: 10.1016/j.intimp.2022.109613] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022]
Abstract
Nuciferine (NCF) is an aporphine alkaloid and a principal bioactive constituent in the lotus plant. Herewith, we investigated the potential anti-inflammatory effect and underlying mechanisms of NCF employing dextran sulfate sodium (DSS)-induced ulcerative colitis in mice, a predominant intestinal inflammatory disease, and mouse RAW 264.7 cells in vitro. Lipopolysaccharide (LPS) was used to generate an inflammatory response in the RAW 264.7 cells. The disease activity index (DAI), colon morphology, colonoscopy, and colon histopathology were performed to assess experimental colitis. The biochemical assays, enzyme-linked immunosorbent assay (ELISA), and immunoblot analysis were performed to understand the underlying mechanisms. In RAW 264.7 cells, NCF pretreatment significantly decreased the expression of inducible nitric oxide synthase (iNOS), the expression and release of pro-inflammatory cytokines including interleukin (IL)-1β, IL-18, and tumor necrosis factor-α (TNF-α) and interfered with the activation of mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB), and NOD-like family pyrin domain containing 3 (NLRP3) signaling pathways. The oral treatment of NCF substantially alleviated the DSS-induced DAI, increased colon length, and restored colon morphology and histology. Compared to the DSS-induced mice, the proteins involved in the activation of MAPK/NF-κB/NLRP3 pathways and the cytokines were markedly decreased in the NCF-treated mice. Moreover, the tight junction architecture of the colon was well-maintained in NCF treatment groups by regulating the expression of claudin-1 and zonula occludens-1 (ZO-1) proteins. All these findings suggest that NCF can be a promising molecule to modulate ulcerative colitis.
Collapse
Affiliation(s)
- Uttam Kulhari
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, 781101 Assam, India
| | - Sourav Kundu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, 781101 Assam, India
| | - Madhav Nilakanth Mugale
- Toxicology & Experimental Medicine, CSIR-Central Drug Research Institute (CDRI), Lucknow 226031, India
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari, 781101 Assam, India.
| |
Collapse
|
7
|
Diminazene aceturate attenuates hepatic ischemia/reperfusion injury in mice. Sci Rep 2022; 12:18158. [PMID: 36307457 PMCID: PMC9616812 DOI: 10.1038/s41598-022-21865-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/04/2022] [Indexed: 12/31/2022] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury is one of the leading causes of mortality following partial hepatectomy, liver transplantation, hypovolemic shock and trauma; however, effective therapeutic targets for the treatment of hepatic I/R injury are lacking. Recent studies have shown that diminazene aceturate (DIZE) has protective effects against inflammation, oxidative stress and cell death, which are the main pathogenetic mechanisms associated with hepatic I/R injury. However, the mechanistic effects DIZE exerts on hepatic I/R remain unknown. C57BL/6 male mice were pretreated with either 15 mg/kg DIZE or vehicle control (saline) and subjected to partial liver ischemia for 60 min. One day after induction of hepatic I/R, liver damage, inflammatory responses, oxidative stress and apoptosis were analyzed. By evaluating plasma alanine aminotransferase levels and histology, we found that DIZE treatment attenuated liver failure and was associated with a reduction in histologically-apparent liver damage. We also found that DIZE-treated mice had milder inflammatory responses, less reactive oxidative damage and less apoptosis following hepatic I/R compared to vehicle-treated mice. Taken together, our study demonstrates that DIZE protects against ischemic liver injury by attenuating inflammation and oxidative damage and may be a potential therapeutic agent for the prevention and treatment of ischemic liver failure.
Collapse
|
8
|
Kumari P, Kumar S, Sethy M, Bhue S, Mohanta BK, Dixit A. Identification of therapeutically potential targets and their ligands for the treatment of OSCC. Front Oncol 2022; 12:910494. [PMID: 36203433 PMCID: PMC9530560 DOI: 10.3389/fonc.2022.910494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Recent advancements in cancer biology have revealed molecular changes associated with carcinogenesis and chemotherapeutic exposure. The available information is being gainfully utilized to develop therapies targeting specific molecules involved in cancer cell growth, survival, and chemoresistance. Targeted therapies have dramatically increased overall survival (OS) in many cancers. Therefore, developing such targeted therapies against oral squamous cell carcinoma (OSCC) is anticipated to have significant clinical implications. In the current work, we have identified drug-specific sensitivity-related prognostic biomarkers (BOP1, CCNA2, CKS2, PLAU, and SERPINE1) using gene expression, Cox proportional hazards regression, and machine learning in OSCC. Dysregulation of these markers is significantly associated with OS in many cancers. Their elevated expression is related to cellular proliferation and aggressive malignancy in various cancers. Mechanistically, inhibition of these biomarkers should significantly reduce cellular proliferation and metastasis in OSCC and should result in better OS. It is pertinent to note that no effective small-molecule candidate has been identified against these biomarkers to date. Therefore, a comprehensive in silico drug design strategy assimilating homology modeling, extensive molecular dynamics (MD) simulation, and ensemble molecular docking has been applied to identify potential compounds against identified targets, and potential molecules have been identified. We hope that this study will help in deciphering potential genes having roles in chemoresistance and a significant impact on OS. It will also result in the identification of new targeted therapeutics against OSCC.
Collapse
Affiliation(s)
- Pratima Kumari
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Sugandh Kumar
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | - Madhusmita Sethy
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | - Shyamlal Bhue
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Bineet Kumar Mohanta
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Anshuman Dixit
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- *Correspondence: Anshuman Dixit,
| |
Collapse
|
9
|
Santos ES, Silva PC, Sousa PSA, Aquino CC, Pacheco G, Teixeira LFLS, Araujo AR, Sousa FBM, Barros RO, Ramos RM, Rocha JA, Nicolau LAD, Medeiros JVR. Antiviral potential of diminazene aceturate against SARS-CoV-2 proteases using computational and in vitro approaches. Chem Biol Interact 2022; 367:110161. [PMID: 36116513 PMCID: PMC9476334 DOI: 10.1016/j.cbi.2022.110161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/05/2022]
Abstract
Diminazene aceturate (DIZE), an antiparasitic, is an ACE2 activator, and studies show that activators of this enzyme may be beneficial for COVID-19, disease caused by SARS-CoV-2. Thus, the objective was to evaluate the in silico and in vitro affinity of diminazene aceturate against molecular targets of SARS-CoV-2. 3D structures from DIZE and the proteases from SARS-CoV-2, obtained through the Protein Data Bank and Drug Database (Drubank), and processed in computer programs like AutodockTools, LigPlot, Pymol for molecular docking and visualization and GROMACS was used to perform molecular dynamics. The results demonstrate that DIZE could interact with all tested targets, and the best binding energies were obtained from the interaction of Protein S (closed conformation −7.87 kcal/mol) and Mpro (−6.23 kcal/mol), indicating that it can act both by preventing entry and viral replication. The results of molecular dynamics demonstrate that DIZE was able to promote a change in stability at the cleavage sites between S1 and S2, which could prevent binding to ACE2 and fusion with the membrane. In addition, in vitro tests confirm the in silico results showing that DIZE could inhibit the binding between the spike receptor-binding domain protein and ACE2, which could promote a reduction in the virus infection. However, tests in other experimental models with in vivo approaches are needed.
Collapse
Affiliation(s)
- Esley S Santos
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil
| | - Priscila C Silva
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Paulo S A Sousa
- Laboratory of Medicinal Chemistry and Biotechnology, QUIMEBIO, Federal University of Maranhão, São Bernardo, MA, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Cristhyane C Aquino
- Postgraduate Program in Medical Sciences, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Gabriella Pacheco
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil
| | - Luiz F L S Teixeira
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Alyne R Araujo
- Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Francisca B M Sousa
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Romulo O Barros
- Research Laboratory in Information Systems, Department of Information, Environment, Health and Food Production, Federal Institute of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Ricardo M Ramos
- Research Laboratory in Information Systems, Department of Information, Environment, Health and Food Production, Federal Institute of Piauí, LaPeSI/IFPI, Teresina, Piauí, Brazil
| | - Jefferson A Rocha
- Laboratory of Medicinal Chemistry and Biotechnology, QUIMEBIO, Federal University of Maranhão, São Bernardo, MA, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Lucas A D Nicolau
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil
| | - Jand V R Medeiros
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil; Medicinal Plants Research Center (NPPM), Federal University of Piauí, Teresina, Brazil; Biodiversity and Biotechnology Research Center, BIOTEC, Post-graduation Program in Biotechnology, Parnaíba Delta Federal University, Parnaíba, PI, Brazil.
| |
Collapse
|
10
|
Lu Z, Wu D, Wang Z, Zhang H, Du Y, Wang G. Diminazene aceturate mitigates cardiomyopathy by interfering with renin-angiotensin system in a septic rat model. BMC Pharmacol Toxicol 2022; 23:44. [PMID: 35787308 PMCID: PMC9251020 DOI: 10.1186/s40360-022-00584-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 06/27/2022] [Indexed: 11/19/2022] Open
Abstract
Background There were limited studies investigating treatments of septic cardiomyopathy (SCM), which is a common complication during sepsis. A septic rat model created by cecal ligation and puncture (CLP) was used to investigate the effects of diminazene aceturate (DIZE) in SCM. Methods A total of 151 Wistar rats were randomly assigned into the sham, CLP, or CLP + DIZE group. Data evaluated postoperatively at 6, 12, 24, and 48 hours included: cardiac function; plasma concentrations of tumor necrosis factor-alpha (TNF-α), interleukin-6, angiotensin-(1–7) [Ang-(1–7)], angiotensin II (AngII), troponin I, and brain natriuretic peptide; expression levels of myocardial Ang-(1–7), angiotensin-converting enzyme (ACE), ACE2, and angiotensin type 1 and Mas receptors; and histological changes. Results We found that the CLP + DIZE group had a lower mortality compared to the CLP group (38.5% versus 61.5%) within 48 h postoperatively, although without statistical significance. In contrast to the sham group, the CLP group had decreased cardiac functions, increased myocardial injuries, and higher TNF-α levels, which were ameliorated in the CLP + DIZE group. Furthermore, administration of DIZE could reverse the decreases of myocardial Ang-(1–7) and ACE2 expressions in the CLP group, which finally minimized the myocardial microstructure disruptions. Conclusions It was concluded that DIZE could mitigate the development of SCM and preserve cardiac function during sepsis possibly by interfering with the renin-angiotensin system through promoting myocardial ACE2 expression and restoring local Ang-(1–7) levels. Supplementary Information The online version contains supplementary material available at 10.1186/s40360-022-00584-4.
Collapse
Affiliation(s)
- Zhaoqing Lu
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Di Wu
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Zheng Wang
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Hanyu Zhang
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Yufan Du
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Guoxing Wang
- Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
11
|
Venturelli A, Tagliazucchi L, Lima C, Venuti F, Malpezzi G, Magoulas GE, Santarem N, Calogeropoulou T, Cordeiro-da-Silva A, Costi MP. Current Treatments to Control African Trypanosomiasis and One Health Perspective. Microorganisms 2022; 10:microorganisms10071298. [PMID: 35889018 PMCID: PMC9321528 DOI: 10.3390/microorganisms10071298] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Human African Trypanosomiasis (HAT, sleeping sickness) and Animal African Trypanosomiasis (AAT) are neglected tropical diseases generally caused by the same etiological agent, Trypanosoma brucei. Despite important advances in the reduction or disappearance of HAT cases, AAT represents a risky reservoir of the infections. There is a strong need to control AAT, as is claimed by the European Commission in a recent document on the reservation of antimicrobials for human use. Control of AAT is considered part of the One Health approach established by the FAO program against African Trypanosomiasis. Under the umbrella of the One Health concepts, in this work, by analyzing the pharmacological properties of the therapeutic options against Trypanosoma brucei spp., we underline the need for clearer and more defined guidelines in the employment of drugs designed for HAT and AAT. Essential requirements are addressed to meet the challenge of drug use and drug resistance development. This approach shall avoid inter-species cross-resistance phenomena and retain drugs therapeutic activity.
Collapse
Affiliation(s)
- Alberto Venturelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - Lorenzo Tagliazucchi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
- Doctorate School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Clara Lima
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Federica Venuti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - Giulia Malpezzi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - George E. Magoulas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (G.E.M.); (T.C.)
| | - Nuno Santarem
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Theodora Calogeropoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (G.E.M.); (T.C.)
| | - Anabela Cordeiro-da-Silva
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
- Correspondence:
| |
Collapse
|
12
|
El-Domiaty HF, Sweed E, Kora MA, Zaki NG, Khodir SA. Activation of angiotensin-converting enzyme 2 ameliorates metabolic syndrome-induced renal damage in rats by renal TLR4 and nuclear transcription factor κB downregulation. Front Med (Lausanne) 2022; 9:904756. [PMID: 36035416 PMCID: PMC9411523 DOI: 10.3389/fmed.2022.904756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) is an independent risk factor for chronic kidney disease (CKD) through many mechanisms, including activation of the renin-angiotensin system. The deleterious effects of angiotensin II (Ang II) can be counterbalanced by angiotensin-converting enzyme 2 (ACE2). Diminazene aceturate (DIZE), an anti-trypanosomal drug, can activate ACE2. OBJECTIVE This study aimed to investigate the possible reno-protective effects of DIZE in MetS rats with elucidation of related mechanisms. MATERIALS AND METHODS Thirty adult male Wistar albino rats were divided equally into control, MetS, and MetS + DIZE groups. Body weight, systolic blood pressure (SBP), and urinary albumin levels were measured. Serum levels of fasting blood glucose (FBG), insulin, uric acid, lipid profile, urea, and creatinine were measured. Homeostasis Model Assessment Index (HOMA-IR) was estimated. Subsequently, renal levels of ACE2, Ang II, malondialdehyde (MDA), reduced glutathione (GSH), and tumor necrosis factor-α (TNF-α) were measured with histopathological and immunohistochemical assessment of TLR4 and NF-κB in renal tissues. RESULTS MetS caused dyslipidemia with significant increases in body weight, SBP, FBG, serum insulin, HOMA-IR, uric acid, urea, creatinine, urinary albumin, and renal levels of Ang II, MDA, and TNF-α, whereas renal ACE2 and GSH were significantly decreased. Renal TLR4 and NF-κB immunoreactivity in MetS rats was upregulated. DIZE supplementation of MetS rats induced significant improvements in renal function parameters; this could be explained by the ability of DIZE to activate renal ACE2 and decrease renal Ang II levels with downregulation of renal TLR4 and NF-κB expression. CONCLUSION DIZE exerts a reno-protective effect in MetS, mainly by downregulating renal TLR4 and NF-κB levels.
Collapse
Affiliation(s)
- Heba F. El-Domiaty
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Eman Sweed
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
- *Correspondence: Eman Sweed,
| | - Mona A. Kora
- Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Nader G. Zaki
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Suzan A. Khodir
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| |
Collapse
|
13
|
Faria BCD, Sacramento LGG, Filipin CSA, da Cruz AF, Nagata SN, Silva ACSE. An analysis of chronic kidney disease as a prognostic factor in pediatric cases of COVID-19. J Bras Nefrol 2021; 43:400-409. [PMID: 33704348 PMCID: PMC8428649 DOI: 10.1590/2175-8239-jbn-2020-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/17/2020] [Indexed: 11/24/2022] Open
Abstract
Advanced age is a risk factor for severe infection by acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Children, however, often present with milder manifestations of Coronavirus Disease 2019 (COVID-19). Associations have been found between COVID-19 and multisystem inflammatory syndrome in children (MIS-C). Patients with the latter condition present more severe involvement. Adults with comorbidities such as chronic kidney disease (CKD) are more severely affected. This narrative review aimed to look into whether CKD contributed to more severe involvement in pediatric patients with COVID-19. The studies included in this review did not report severe cases or deaths, and indicated that pediatric patients with CKD and previously healthy children recovered quickly from infection. However, some patients with MIS-C required hospitalization in intensive care units and a few died, although it was not possible to correlate MIS-C and CKD. Conversely, adults with CKD reportedly had increased risk of severe infection by SARS-CoV-2 and higher death rates. The discrepancies seen between age groups may be due to immune system and renin-angiotensin system differences, with more pronounced expression of ACE2 in children. Immunosuppressant therapy has not been related with positive or negative effects in individuals with COVID-19, although current recommendations establish decreases in the dosage of some medications. To sum up with, CKD was not associated with more severe involvement in children diagnosed with COVID-19. Studies enrolling larger populations are still required.
Collapse
Affiliation(s)
| | | | | | - Aniel Feitosa da Cruz
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo
Horizonte, MG, Brasil
| | - Sarah Naomi Nagata
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo
Horizonte, MG, Brasil
| | - Ana Cristina Simões e Silva
- Universidade Federal de Minas Gerais, Faculdade de Medicina,
Departamento de Pediatria, Belo Horizonte, MG, Brasil
| |
Collapse
|
14
|
Pantazi I, Al-Qahtani AA, Alhamlan FS, Alothaid H, Matou-Nasri S, Sourvinos G, Vergadi E, Tsatsanis C. SARS-CoV-2/ACE2 Interaction Suppresses IRAK-M Expression and Promotes Pro-Inflammatory Cytokine Production in Macrophages. Front Immunol 2021; 12:683800. [PMID: 34248968 PMCID: PMC8261299 DOI: 10.3389/fimmu.2021.683800] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/31/2021] [Indexed: 01/08/2023] Open
Abstract
The major cause of death in SARS-CoV-2 infected patients is due to de-regulation of the innate immune system and development of cytokine storm. SARS-CoV-2 infects multiple cell types in the lung, including macrophages, by engagement of its spike (S) protein on angiotensin converting enzyme 2 (ACE2) receptor. ACE2 receptor initiates signals in macrophages that modulate their activation, including production of cytokines and chemokines. IL-1R-associated kinase (IRAK)-M is a central regulator of inflammatory responses regulating the magnitude of TLR responsiveness. Aim of the work was to investigate whether SARS-CoV-2 S protein-initiated signals modulate pro-inflammatory cytokine production in macrophages. For this purpose, we treated PMA-differentiated THP-1 human macrophages with SARS-CoV-2 S protein and measured the induction of inflammatory mediators including IL6, TNFα, IL8, CXCL5, and MIP1a. The results showed that SARS-CoV-2 S protein induced IL6, MIP1a and TNFα mRNA expression, while it had no effect on IL8 and CXCL5 mRNA levels. We further examined whether SARS-CoV-2 S protein altered the responsiveness of macrophages to TLR signals. Treatment of LPS-activated macrophages with SARS-CoV-2 S protein augmented IL6 and MIP1a mRNA, an effect that was evident at the protein level only for IL6. Similarly, treatment of PAM3csk4 stimulated macrophages with SARS-CoV-2 S protein resulted in increased mRNA of IL6, while TNFα and MIP1a were unaffected. The results were confirmed in primary human peripheral monocytic cells (PBMCs) and isolated CD14+ monocytes. Macrophage responsiveness to TLR ligands is regulated by IRAK-M, an inactive IRAK kinase isoform. Indeed, we found that SARS-CoV-2 S protein suppressed IRAK-M mRNA and protein expression both in THP1 macrophages and primary human PBMCs and CD14+ monocytes. Engagement of SARS-CoV-2 S protein with ACE2 results in internalization of ACE2 and suppression of its activity. Activation of ACE2 has been previously shown to induce anti-inflammatory responses in macrophages. Treatment of macrophages with the ACE2 activator DIZE suppressed the pro-inflammatory action of SARS-CoV-2. Our results demonstrated that SARS-CoV-2/ACE2 interaction rendered macrophages hyper-responsive to TLR signals, suppressed IRAK-M and promoted pro-inflammatory cytokine expression. Thus, activation of ACE2 may be a potential anti-inflammatory therapeutic strategy to eliminate the development of cytokine storm observed in COVID-19 patients.
Collapse
Affiliation(s)
- Ioanna Pantazi
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece.,Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece
| | - Ahmed A Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Fatimah S Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hani Alothaid
- Department of Basic Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Sabine Matou-Nasri
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - George Sourvinos
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Greece
| | - Eleni Vergadi
- Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| |
Collapse
|
15
|
Stachowicz A, Wiśniewska A, Kuś K, Białas M, Łomnicka M, Totoń-Żurańska J, Kiepura A, Stachyra K, Suski M, Bujak-Giżycka B, Jawień J, Olszanecki R. Diminazene Aceturate Stabilizes Atherosclerotic Plaque and Attenuates Hepatic Steatosis in apoE-Knockout Mice by Influencing Macrophages Polarization and Taurine Biosynthesis. Int J Mol Sci 2021; 22:5861. [PMID: 34070749 PMCID: PMC8199145 DOI: 10.3390/ijms22115861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis and nonalcoholic fatty liver disease are leading causes of morbidity and mortality in the Western countries. The renin-angiotensin system (RAS) with its two main opposing effectors, i.e., angiotensin II (Ang II) and Ang-(1-7), is widely recognized as a major regulator of cardiovascular function and body metabolic processes. Angiotensin-converting enzyme 2 (ACE2) by breaking-down Ang II forms Ang-(1-7) and thus favors Ang-(1-7) actions. Therefore, the aim of our study was to comprehensively evaluate the influence of prolonged treatment with ACE2 activator, diminazene aceturate (DIZE) on the development of atherosclerotic lesions and hepatic steatosis in apoE-/- mice fed a high-fat diet (HFD). We have shown that DIZE stabilized atherosclerotic lesions and attenuated hepatic steatosis in apoE-/- mice fed an HFD. Such effects were associated with decreased total macrophages content and increased α-smooth muscle actin levels in atherosclerotic plaques. Moreover, DIZE changed polarization of macrophages towards increased amount of anti-inflammatory M2 macrophages in the atherosclerotic lesions. Interestingly, the anti-steatotic action of DIZE in the liver was related to the elevated levels of HDL in the plasma, decreased levels of triglycerides, and increased biosynthesis and concentration of taurine in the liver of apoE-/- mice. However, exact molecular mechanisms of both anti-atherosclerotic and anti-steatotic actions of DIZE require further investigations.
Collapse
Affiliation(s)
- Aneta Stachowicz
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Katarzyna Kuś
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Magdalena Białas
- Chair of Pathomorphology, Jagiellonian University Medical College, 31-531 Krakow, Poland;
| | - Magdalena Łomnicka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Justyna Totoń-Żurańska
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Anna Kiepura
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Kamila Stachyra
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Maciej Suski
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Beata Bujak-Giżycka
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Jacek Jawień
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| | - Rafał Olszanecki
- Chair of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland; (A.W.); (K.K.); (M.Ł.); (J.T.-Ż.); (A.K.); (K.S.); (M.S.); (B.B.-G.); (J.J.); (R.O.)
| |
Collapse
|
16
|
Cytotoxicity and anti-inflammatory effect of a novel diminazene aceturate derivative in bovine mammary epithelial cells. Res Vet Sci 2021; 137:102-110. [PMID: 33964615 DOI: 10.1016/j.rvsc.2021.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 03/23/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
Diminazene aceturate (DA) has been used in the treatment of infections of trypanosomes in animals. Interestingly, its anti-inflammatory effect has recently gained increased interests. However, DA has been reported to have toxic side effects that limit its application. Therefore, we synthesized and screened a novel low-toxic DA derivative, namely the DA derivative 3 (DAD3). In the present study, anti-inflammatory effect of DAD3 was evaluated bovine mammary epithelial cells (BMECs) in vitro model. The results demonstrated that DAD3 had less cytotoxicity, and had a stronger effect in inhibiting secretion of inflammatory factors in BMECs, compared to DA. Mechanistically, DAD3 was able to inhibit the production of pro-inflammatory factors in part by suppressing the generation of mitochondrial reactive oxygen species (ROS) in BMECs upon LPS stimulation. Molecular analysis further indicated that DAD3 was capable of resolving inflammation in BMECs through a mechanism by preventing nuclear translocation of NF-p65, subsequently inhibiting transcription of inflammatory factors. In this context, DAD3 inhibited the phosphorylation of IκB, ERK, JNK and P-38 proteins of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. These results suggested the DAD3 was a novel DA derivative with low toxicity and strong anti-inflammatory effects in BMECs exposed to LPS, through a mechanism by blocking the NF-κB and MAPK signaling pathways. This study also provides an evidence that the DAD3 may be a novel anti-inflammatory agents warranted for further investigation in treatment of mastitis in cows.
Collapse
|
17
|
Kosutova P, Mikolka P, Balentova S, Adamkov M, Calkovska A, Mokra D. Effects of PDE3 Inhibitor Olprinone on the Respiratory Parameters, Inflammation, and Apoptosis in an Experimental Model of Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:E3382. [PMID: 32403267 PMCID: PMC7247002 DOI: 10.3390/ijms21093382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
This study aimed to investigate whether a selective phosphodiesterase-3 (PDE3) inhibitor olprinone can positively influence the inflammation, apoptosis, and respiratory parameters in animals with acute respiratory distress syndrome (ARDS) model induced by repetitive saline lung lavage. Adult rabbits were divided into 3 groups: ARDS without therapy (ARDS), ARDS treated with olprinone i.v. (1 mg/kg; ARDS/PDE3), and healthy ventilated controls (Control), and were oxygen-ventilated for the following 4 h. Dynamic lung-thorax compliance (Cdyn), mean airway pressure (MAP), arterial oxygen saturation (SaO2), alveolar-arterial gradient (AAG), ratio between partial pressure of oxygen in arterial blood to a fraction of inspired oxygen (PaO2/FiO2), oxygenation index (OI), and ventilation efficiency index (VEI) were evaluated every hour. Post mortem, inflammatory and oxidative markers (interleukin (IL)-6, IL-1β, a receptor for advanced glycation end products (RAGE), IL-10, total antioxidant capacity (TAC), 3-nitrotyrosine (3NT), and malondialdehyde (MDA) and apoptosis (apoptotic index and caspase-3) were assessed in the lung tissue. Treatment with olprinone reduced the release of inflammatory mediators and markers of oxidative damage decreased apoptosis of epithelial cells and improved respiratory parameters. The results indicate a future potential of PDE3 inhibitors also in the therapy of ARDS.
Collapse
Affiliation(s)
- Petra Kosutova
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Pavol Mikolka
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Sona Balentova
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (S.B.); (M.A.)
| | - Andrea Calkovska
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| | - Daniela Mokra
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia; (P.K.); (P.M.); (A.C.)
| |
Collapse
|
18
|
Qaradakhi T, Gadanec LK, McSweeney KR, Tacey A, Apostolopoulos V, Levinger I, Rimarova K, Egom EE, Rodrigo L, Kruzliak P, Kubatka P, Zulli A. The potential actions of angiotensin-converting enzyme II (ACE2) activator diminazene aceturate (DIZE) in various diseases. Clin Exp Pharmacol Physiol 2020; 47:751-758. [PMID: 31901211 DOI: 10.1111/1440-1681.13251] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 01/28/2023]
Abstract
The renin angiotensin system (RAS) regulates fluid balance, blood pressure and maintains vascular tone. The potent vasoconstrictor angiotensin II (Ang II) produced by angiotensin-converting enzyme (ACE) comprises the classical RAS. The non-classical RAS involves the conversion of Ang II via ACE2 into the vasodilator Ang (1-7) to counterbalance the effects of Ang II. Furthermore, ACE2 converts AngA into another vasodilator named alamandine. The over activation of the classical RAS (increased vasoconstriction) and depletion of the non-classical RAS (decreased vasodilation) results in vascular dysfunction. Vascular dysfunction is the leading cause of atherosclerosis and cardiovascular disease (CVD). Additionally, local RAS is expressed in various tissues and regulates cellular functions. RAS dysregulation is involved in other several diseases such as inflammation, renal dysfunction and even cancer growth. An approach in restoring vascular dysfunction and other pathological diseases is to either increase the activity of ACE2 or reduce the effect of the classical RAS by counterbalancing Ang II effects. The antitrypanosomal agent, diminazene aceturate (DIZE), is one approach in activating ACE2. DIZE has been shown to exert beneficial effects in CVD experimental models of hypertension, myocardial infarction, type 1 diabetes and atherosclerosis. Thus, this review focuses on DIZE and its effect in several tissues such as blood vessels, cardiac, renal, immune and cancer cells.
Collapse
Affiliation(s)
- Tawar Qaradakhi
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | | | - Alexander Tacey
- Institute for Health and Sport, Victoria University, Melbourne, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, Australia
| | | | - Itamar Levinger
- Institute for Health and Sport, Victoria University, Melbourne, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, Australia
| | - Kvetoslava Rimarova
- Department of Public Health and Hygiene, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Emmanuel E Egom
- Egom Clinical & Translational Research Services Ltd, Dartmouth, NS, Canada.,Jewish General Hospital and Lady Davis Research Institute, Montreal, QC, Canada
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo and Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Peter Kruzliak
- Department of Internal Medicine, Borthers of Mercy Hospital, Brno, Czech Republic.,2nd Department of Surgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia.,Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| |
Collapse
|
19
|
Nemoto W, Yamagata R, Nakagawasai O, Nakagawa K, Hung WY, Fujita M, Tadano T, Tan-No K. Effect of spinal angiotensin-converting enzyme 2 activation on the formalin-induced nociceptive response in mice. Eur J Pharmacol 2020; 872:172950. [PMID: 31987711 DOI: 10.1016/j.ejphar.2020.172950] [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: 07/24/2019] [Revised: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 12/30/2022]
Abstract
We have previously demonstrated that the phosphorylation of p38 MAPK, through spinal AT1 receptor activation, is involved in formalin-induced nociception and follows accompanied by the increase in spinal angiotensin (Ang) II levels. We have also found that Ang (1-7), an N-terminal fragment of Ang II generated by ACE2, prevents the Ang II-induced nociceptive behavior via spinal MAS1 and the inhibition of p38 MAPK phosphorylation. Here, we examined whether the ACE2 activator diminazene aceturate (DIZE) can prevent the formalin-induced nociception in mice. The i.t. administration of DIZE attenuated the second, but not the first phase of formalin-induced nociceptive response. An increase in the activity of spinal ACE2 was measured following DIZE administration. The inhibitory effect of DIZE on nociception was abolished by the i.t. co-administration of the MAS1 antagonist A779. The i.t. administration of Ang (1-7) showed a similar effect on the second phase of the response which was also attenuated by A779. Furthermore, DIZE and Ang (1-7) each inhibited the formalin-induced phosphorylation of p38 MAPK on the dorsal lumbar spinal cord. This inhibition was again prevented by A779. ACE2 was expressed in neurons and microglia but absent from astrocytes in the superficial dorsal horn. Our data show that the i.t.-administered DIZE attenuates the second phase of the formalin-induced nociception which is accompanied by the inhibition of p38 MAPK phosphorylation. They also suggest the involvement of MAS1 activation on spinal neurons and microglia in response to the increase in Ang (1-7) following ACE2 activation.
Collapse
Affiliation(s)
- Wataru Nemoto
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan.
| | - Ryota Yamagata
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan
| | - Osamu Nakagawasai
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan
| | - Koharu Nakagawa
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan
| | - Wan-Yi Hung
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan
| | - Maho Fujita
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan
| | - Takeshi Tadano
- Complementary and Alternative Medicine Clinical Research and Development, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Koichi Tan-No
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, 981-8558, Japan
| |
Collapse
|
20
|
Yang J, Chen Y, Jiang K, Yang Y, Zhao G, Guo S, Deng G. MicroRNA-106a Provides Negative Feedback Regulation in Lipopolysaccharide-Induced Inflammation by targeting TLR4. Int J Biol Sci 2019; 15:2308-2319. [PMID: 31595149 PMCID: PMC6775322 DOI: 10.7150/ijbs.33432] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/03/2019] [Indexed: 12/13/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical disease with high incidence and mortality rate, which is characterized by severe inflammatory response and tissues damage. MicroRNAs (miRNAs) have been regarded as novel regulators of inflammation, and play an important role in various inflammatory diseases. However, it remains unknown whether the regulatory mechanisms mediated by miR-106a is involved in LPS-induced ALI. In this study, we found that expression of miR-106a was significantly decreased in lung tissues of ALI mice and LPS-stimulated macrophages. We also revealed that over-expression of miR-106a significantly decreased the production of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α, whereas this effect was reversed by the inhibition of miR-106a. Moreover, miR-106a inhibits NF-κB activation by targeting TLR4 expression. We further demonstrated that miR-106a inhibited TLR4 expression via binding directly to the 3'-UTR of TLR4. Taken together, the results of the present study illuminated that miR-106a is a negative feedback regulator in LPS-stimulated inflammation through TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China. College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| |
Collapse
|
21
|
Glycitin alleviates lipopolysaccharide-induced acute lung injury via inhibiting NF-κB and MAPKs pathway activation in mice. Int Immunopharmacol 2019; 75:105749. [PMID: 31306981 DOI: 10.1016/j.intimp.2019.105749] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a pulmonary diffuse dysfunction disease caused by immoderate inflammatory response breaking the coordination of physiological structures and functions, and there are very few effective treatments to reduce high morbidity of ALI in critical patients. Glycitin is a natural ingredient derived from the seeds of leguminous plants and may have potent anti-inflammation features. The purpose of this study was to investigate the anti-inflammation effect of glycitin on LPS-induced ALI in mice and elucidate its possible anti-inflammatory mechanisms. The results of histopathological changes, the wet/dry weight ratio as well as the myeloperoxidase (MPO) activity indicated that glycitin obviously alleviated the lung injury induced by LPS. In addition, qPCR and ELISA results found that glycitin could dose-dependently decrease the expressions of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α. Western blotting was performed to revealed that glycitin inhibited the activation of NF-κB and MAPKs signaling pathways by suppressing the expression of TLR4 protein and the phosphorylation of IKKβ, IκBα, p65, p38, ERK, and JNK. All data indicated that glycitin could protect lung tissues from LPS-induced inflammation via inhibiting TLR4-mediated NF-κB and MAPKs signaling pathways.
Collapse
|
22
|
Abstract
The plant Zanthoxylum zanthoxyloides (Lam.) Zepern. & Timler is one of the most important medicinal species of the genus Zanthoxylum on the African continent. It is used in the treatment and management of parasitic diseases in sub-Saharan Africa. These properties have inspired scientists to investigate species within the genus for bioactive compounds. However, a study, which details a spectroscopic, spectrometric and bioactivity guided extraction and isolation of antiparasitic compounds from the genus Zanthoxylum is currently non-existent. Tortozanthoxylamide (1), which is a derivative of the known compound armatamide was isolated from Z. zanthoxyloides and the full structure determined using UV, IR, 1D/2D-NMR and high-resolution liquid chromatography tandem mass spectrometry (HRESI-LC-MS) data. When tested against Trypanosoma brucei subsp. brucei, the parasite responsible for animal African trypanosomiasis in sub-Saharan Africa, 1 (IC50 7.78 µM) was just four times less active than the commercially available drug diminazene aceturate (IC50 1.88 µM). Diminazene aceturate is a potent drug for the treatment of animal African trypanosomiasis. Tortozanthoxylamide (1) exhibits a significant antitrypanosomal activity through remarkable alteration of the cell cycle in T. brucei subsp. brucei, but it is selectively non-toxic to mouse macrophages RAW 264.7 cell lines. This suggests that 1 may be considered as a scaffold for the further development of natural antitrypanosomal compounds.
Collapse
|
23
|
Guo S, Chen Y, Liu J, Yang J, Yang C, Zhang T, Jiang K, Wu Z, Shaukat A, Deng G. miR-497a-5p attenuates lipopolysaccharide-induced inflammatory injury by targeting IRAK2. J Cell Physiol 2019; 234:22874-22883. [PMID: 31148190 DOI: 10.1002/jcp.28850] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022]
Abstract
Acute lung injury (ALI) is a severe acute inflammatory reaction of the lungs caused by a variety of factors, which can lead to a high mortality rate. MicroRNAs are a novel therapeutic molecule that play a vital role in many diseases. However, its mechanism of action in lipopolysaccharide (LPS)-induced mouse ALI is not clear. The study aimed to investigate the mechanism of action of miR-497 in LPS-induced ALI. As a result, it was found that the expression of miR-497 in the inflammatory reaction showed a decrease in time and dose trends. Importantly, miR-497 reduced LPS-induced expression levels of related inflammatory factors. In addition, we also demonstrated that IRAK2 is a direct target molecule of miR-497. Interestingly, we further found that miR-497 inhibits the expression of IRAK2 by targeting IRAK2-3'UTR. Therefore, miR-497 can partially negatively regulate the activation of IRAK2-NF-κB pathway in LPS-induced inflammatory responses.
Collapse
Affiliation(s)
- Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Junfeng Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Chao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Zhimin Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
24
|
Ginsenoside Rb1 ameliorates Staphylococcus aureus-induced Acute Lung Injury through attenuating NF-κB and MAPK activation. Microb Pathog 2019; 132:302-312. [PMID: 31059756 DOI: 10.1016/j.micpath.2019.05.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 11/24/2022]
Abstract
Acute lung injury (ALI) is clinically characterized by excessive inflammation leading to acute respiratory distress syndrome (ARDS), having high morbidity and mortality both in human and animals. Ginsenoside Rb1 (Rb1) is a major primary bioactive component extracted by Panax ginseng, which has numerous pharmacological functions such as anti-cancer, anti-inflammatory, and antioxidant. However, the anti-inflammatory effects of Rb1 in Staphylococcus aureus (S. aureus)-induced ALI in mice have not been investigated. The aim of the current study was to determine the anti-inflammatory influence of Rb1 on S. aureus-induced ALI in mice, and to explore its possible underlying principle mechanisms in RAW 264.7 macrophage cells. The results of physical morphology, histopathological variation and wet-to-dry weight ratio of lungs revealed that Rb1 significantly attenuated S. aureus-induced lung injury. Furthermore, qPCR results displayed that Rb1 inhibited IL-1β, IL-6 and TNF-α production both in vivo and in vitro. The activation of Toll-like receptor 2 (TLR2) by S. aureus was inhibited by application of Rb1 as confirmed by results of immunofluorescence assay. The expression of NF-kB and MAPK signaling proteins revealed that Rb1 significantly attenuated the phosphorylation of p65, ERK, as well as JNK. Altogether, the results of this experiment presented that Rb1 has ability to protect S. aureus-induced ALI in mice by attenuating TLR-2-mediated NF-kB and MAPK signaling pathways. Consequently, Rb-1 might be a potential medicine in the treatment of S. aureus-induced lung inflammation.
Collapse
|
25
|
Diminazene aceturate (Berenil) downregulates Trypanosoma congolense-induced proinflammatory cytokine production by altering phosphorylation of MAPK and STAT proteins. Immunol Res 2018; 67:84-92. [DOI: 10.1007/s12026-018-9040-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
26
|
Guo S, Jiang K, Wu H, Yang C, Yang Y, Yang J, Zhao G, Deng G. Magnoflorine Ameliorates Lipopolysaccharide-Induced Acute Lung Injury via Suppressing NF-κB and MAPK Activation. Front Pharmacol 2018; 9:982. [PMID: 30214410 PMCID: PMC6125611 DOI: 10.3389/fphar.2018.00982] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/10/2018] [Indexed: 01/27/2023] Open
Abstract
Acute lung injury (ALI) which is featured by a strong pulmonary inflammation, is a major cause of morbidity and mortality in critically ill patients. Magnoflorine, a quaternary alkaloid isolated from Chinese herb Magnolia or Aristolochia, has been reported to have potent anti-inflammatory properties. However, the effect of magnoflorine on lipopolysaccharide (LPS)-induced ALI in mice has not been reported. The purpose of the present study is to investigate the anti-inflammatory effect of magnoflorine on LPS-induced ALI and elucidate its possible molecular mechanisms in RAW264.7 cells. The results of histopathological changes as well as the myeloperoxidase (MPO) activity indicated that magnoflorine significantly alleviated the lung injury induced by LPS. In addition, qPCR results showed that magnoflorine dose-dependently decreased the expression of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6. Immunofluorescence assay also confirmed that the level of Toll-like receptor 4 (TLR4) induced by LPS was inhibited by magnoflorine treatment. Further experiments were performed using Western blotting to detect the expression of related proteins in the NF-κB and MAPK signaling pathways. The results showed that magnoflorine suppressed the levels of phosphorylated p65, IκBα, p38, ERK, and JNK. In conclusion, all data indicate that magnoflorine could protect against LPS-induced inflammation in ALI at least partially by inhibiting TLR4-mediated NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
27
|
Ge P, Yao X, Li J, Jiang R, Dai J, Zhang L. Diminazene aceturate alleviated lipopolysaccharide/D-galactosamine-induced fulminant hepatitis in mice. Biomed Pharmacother 2018; 98:142-148. [DOI: 10.1016/j.biopha.2017.12.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
|
28
|
Lee JYP, Saez NJ, Cristofori-Armstrong B, Anangi R, King GF, Smith MT, Rash LD. Inhibition of acid-sensing ion channels by diminazene and APETx2 evoke partial and highly variable antihyperalgesia in a rat model of inflammatory pain. Br J Pharmacol 2018; 175:2204-2218. [PMID: 29134638 DOI: 10.1111/bph.14089] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/22/2017] [Accepted: 10/31/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Acid-sensing ion channels (ASICs) are primary acid sensors in mammals, with the ASIC1b and ASIC3 subtypes being involved in peripheral nociception. The antiprotozoal drug diminazene is a moderately potent ASIC inhibitor, but its analgesic activity has not been assessed. EXPERIMENTAL APPROACH We determined the ASIC subtype selectivity of diminazene and the mechanism by which it inhibits ASICs using voltage-clamp electrophysiology of Xenopus oocytes expressing ASICs 1-3. Its peripheral analgesic activity was then assessed relative to APETx2, an ASIC3 inhibitor, and morphine, in a Freund's complete adjuvant (FCA)-induced rat model of inflammatory pain. KEY RESULTS Diminazene inhibited homomeric rat ASICs with IC50 values of ~200-800 nM, via an open channel and subtype-dependent mechanism. In rats with FCA-induced inflammatory pain in one hindpaw, diminazene and APETx2 evoked more potent peripheral antihyperalgesia than morphine, but the effect was partial for APETx2. APETx2 potentiated rat ASIC1b at concentrations 30-fold to 100-fold higher than the concentration inhibiting ASIC3, which may have implications for its use in in vivo experiments. CONCLUSIONS AND IMPLICATIONS Diminazene and APETx2 are moderately potent ASIC inhibitors, both inducing peripheral antihyperalgesia in a rat model of chronic inflammatory pain. APETx2 has a more complex ASIC pharmacology, which must be considered when it is used as a supposedly selective ASIC3 inhibitor in vivo. Our use of outbred rats revealed responders and non-responders when ASIC inhibition was used to alleviate inflammatory pain, which is aligned with the concept of number-needed-to-treat in human clinical studies. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Jia Yu Peppermint Lee
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia
| | - Natalie J Saez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | | | - Raveendra Anangi
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Maree T Smith
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, St Lucia, QLD, Australia
| | - Lachlan D Rash
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
29
|
Wu H, Yang Y, Guo S, Yang J, Jiang K, Zhao G, Qiu C, Deng G. Nuciferine Ameliorates Inflammatory Responses by Inhibiting the TLR4-Mediated Pathway in Lipopolysaccharide-Induced Acute Lung Injury. Front Pharmacol 2017; 8:939. [PMID: 29311940 PMCID: PMC5742629 DOI: 10.3389/fphar.2017.00939] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022] Open
Abstract
Acute lung injury (ALI) is a complex syndrome with sepsis occurring in critical patients, who usually lack effective therapy. Nuciferine is a primary bioactive component extracted from the lotus leaf, and it displays extensive pharmacological functions, including anti-cancer, anti-inflammatory, and antioxidant properties. Nevertheless, the effects of nuciferine on lipopolysaccharide (LPS)-stimulated ALI in mice has not been investigated. ALI of mice stimulated by LPS was used to determine the anti-inflammatory function of nuciferine. The molecular mechanism of nuciferine was performed on RAW264.7 macrophage cells. The results of pathological section, myeloperoxidase activity and lung wet/dry ratio showed that nuciferine alleviated LPS-induced lung injury (p < 0.05). qRT-PCR and ELISA experiments suggested that nuciferine inhibited TNF-α, IL-6, and IL-1β secretion in tissues and RAW264.7 cells but increased IL-10 secretion (p < 0.05). Molecular studies showed that TLR4 expression and nuclear factor (NF)-κB activation were both inhibited by nuciferine treatment (p < 0.05). To further investigate the anti-inflammatory mechanism of nuciferine, TLR4 was knocked down. When TLR4 was silenced, LPS induced the production of IL-1β, and TNF-α was markedly decreased by TLR4-siRNA and nuciferine treatment in LPS-induced RAW264.7 cells (p < 0.05). These results suggested that nuciferine had the ability to protect against LPS-stimulated ALI. Thus, nuciferine may be a potential drug for treating LPS-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
30
|
Wu H, Jiang K, Yin N, Ma X, Zhao G, Qiu C, Deng G. Thymol mitigates lipopolysaccharide-induced endometritis by regulating the TLR4- and ROS-mediated NF-κB signaling pathways. Oncotarget 2017; 8:20042-20055. [PMID: 28223539 PMCID: PMC5386742 DOI: 10.18632/oncotarget.15373] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/24/2017] [Indexed: 12/18/2022] Open
Abstract
The purpose of this study was to investigate the effects of thymol on lipopolysaccharide (LPS)-induced inflammatory responses and to clarify the potential mechanism of these effects. LPS-induced mouse endometritis was used to confirm the anti-inflammatory action of thymol in vivo. RAW264.7 cells were used to examine the molecular mechanism and targets of thymol in vitro. In vivo, thymol markedly alleviated LPS-induced pathological injury, myeloperoxidase (MPO) activity, and the production of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in mice. Further studies were performed to examine the expression of the Toll-like receptor 4 (TLR4) -mediated nuclear factor-κB (NF-κB) pathway. These results showed that the expression of the TLR4-mediated NF-κB pathway was inhibited by thymol treatment. In vitro, we observed that thymol dose-dependently inhibited the expression of TNF-α, IL-1β, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) in LPS-stimulated RAW264.7 cells. Moreover, the results obtained from immunofluorescence assays also indicated that thymol dose-dependently suppressed LPS-induced reactive oxygen species (ROS) production. Silencing of TLR4 inhibited NF-κB pathway activation. Furthermore, H2O2 treatment increased the phosphorylation of p65 and IκBα, which were decreased when treated with N-acetyl cysteine or thymol. In conclusion, the anti-inflammatory effects of thymol are associated with activation of the TLR4 or ROS signaling pathways, contributing to NF-κB activation, thereby alleviating LPS-induced oxidative and inflammatory responses.
Collapse
Affiliation(s)
- Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Nannan Yin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Xiaofei Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| |
Collapse
|
31
|
Khadem F, Jia P, Mou Z, Feiz Barazandeh A, Liu D, Keynan Y, Uzonna JE. Pharmacological inhibition of p110δ subunit of PI3K confers protection against experimental leishmaniasis. J Antimicrob Chemother 2016; 72:467-477. [DOI: 10.1093/jac/dkw448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/15/2016] [Accepted: 09/22/2016] [Indexed: 01/17/2023] Open
|
32
|
AAV8-Mediated Angiotensin-Converting Enzyme 2 Gene Delivery Prevents Experimental Autoimmune Uveitis by Regulating MAPK, NF-κB and STAT3 Pathways. Sci Rep 2016; 6:31912. [PMID: 27558087 PMCID: PMC4997264 DOI: 10.1038/srep31912] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/26/2016] [Indexed: 12/27/2022] Open
Abstract
Renin angiotensin system (RAS) is a key hormonal system which regulates the cardiovascular function and is implicated in several autoimmune diseases. With the discovery of the angiotensin-converting enzyme 2 (ACE2), a protective axis of RAS namely ACE2/Ang-(1-7)/Mas that counteracts the deleterious ACE/AngII/AT1R axis has been established. This axis is emerging as a novel target to attenuate ocular inflammation. However, the underlying molecular mechanisms remain unclear. We investigated the hypothesis that enhancing the activity of the protective axis of RAS by subretinal delivery of an AAV8 (Y733F)-ACE2 vector would protect against the ocular inflammation in experimental autoimmune uveitis (EAU) mice through regulating the local immune responses. Our studies demonstrated that increased ACE2 expression exerts protective effects on inflammation in EAU mouse by modulating ocular immune responses, including the differentiation of Th1/Th17 cells and the polarization of M1/M2 macrophages; whereas the systemic immune responses appeared not affected. These effects were mediated by activating the Ang-(1-7)/Mas and inhibiting the MAPK, NF-κB and STAT3 signaling pathways. This proof-of-concept study suggests that activation of ocular ACE2/Ang-(1-7)/Mas axis with AAV gene transfer modulates local immune responses and may be a promising, long-lasting therapeutic strategy for refractory and recurrent uveitis, as well as other inflammatory eye diseases.
Collapse
|
33
|
Kuriakose SM, Singh R, Uzonna JE. Host Intracellular Signaling Events and Pro-inflammatory Cytokine Production in African Trypanosomiasis. Front Immunol 2016; 7:181. [PMID: 27242788 PMCID: PMC4872169 DOI: 10.3389/fimmu.2016.00181] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/27/2016] [Indexed: 12/21/2022] Open
Abstract
Pathogens, such as bacteria, viruses, and parasites, possess specific molecules or proteins that are recognized by several host innate immune receptors, leading to the activation of several intracellular signaling molecules and pathways. The magnitude and quality of these events significantly affect the outcome of infection. African trypanosomes, including Trypanosoma congolense, are capable of manipulating the host immune response, including the activity of macrophages, which are the key immune cells that contribute to the immunopathogenesis of African trypanosomiasis. Although it is known that immune hyperactivation and excessive pro-inflammatory cytokine production are the hallmarks of African trypanosomiasis, the mechanisms through which these events are triggered are poorly defined. However, it is known that macrophages may play a significant role in these processes, because phagocytosis of trypanosomes by macrophages initiates intracellular signal transduction cascades that lead to the release of pro-inflammatory cytokines and alteration in cell function. This review highlights recent progress in our understanding of the innate immune receptors, signaling pathways, and transcription factors involved in T. congolense-induced pro-inflammatory cytokine production in macrophages. It will reveal the existence of complex signaling events through which the parasite modulates the host immune response, thus identifying novel targets that could aid in designing strategies to effectively control the disease.
Collapse
Affiliation(s)
- Shiby M Kuriakose
- Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, MB , Canada
| | - Rani Singh
- Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, MB , Canada
| | - Jude E Uzonna
- Department of Immunology, Faculty of Health Sciences, University of Manitoba , Winnipeg, MB , Canada
| |
Collapse
|
34
|
Tao L, Qiu Y, Fu X, Lin R, Lei C, Wang J, Lei B. Angiotensin-converting enzyme 2 activator diminazene aceturate prevents lipopolysaccharide-induced inflammation by inhibiting MAPK and NF-κB pathways in human retinal pigment epithelium. J Neuroinflammation 2016; 13:35. [PMID: 26862037 PMCID: PMC4748536 DOI: 10.1186/s12974-016-0489-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/20/2016] [Indexed: 01/10/2023] Open
Abstract
Background Retinal inflammation is a devastating pathological process in ocular diseases. Functional impairment of retinal pigment epithelium (RPE) is associated with inflammatory retinal diseases. Enhancing the protective axis namely ACE2/Ang-(1-7)/Mas by activation of ACE2 presents anti-inflammatory properties. We investigated whether diminazene aceturate (DIZE), an angiotensin-converting enzyme 2 (ACE2) activator, prevented lipopolysaccharide (LPS)-induced inflammatory response by activating the protective axis and whether the effect was mediated by inhibiting the mitogen-activated protein kinase (MAPK) and the nuclear factor-κB (NF-κB) pathways. Methods Cell counting kit-8 (CCK-8) assay and real-time PCR were used to determine the optimum concentration and incubation time of DIZE. ARPE-19 cells and primary cultured human retinal pigment epithelia (hRPE) were incubated with or without 10 μg/mL DIZE for 6 h before stimulated with 5 μg/mL LPS for 24 h. The mRNA expression of inflammatory cytokines, AT1R, and AT2R was analyzed. The protein level of inflammatory cytokines, Ang II, and Ang-(1-7) was detected. Phosphorylation of p38 MAPK, extracellular signal-regulated kinase (ERK)1/2, c-Jun N-terminal kinase (JNK) and phosphorylated transcription inhibition factor-κB-α (p-IκB-α) were measured. Inhibitors of MAPKs and NF-κB were added to verify the involvement of these pathways. A small interfering RNA (siRNA) targeted to ACE2 and a selective Ang-(1-7) antagonist A779 was used to confirm the role of ACE2 and the involvement of ACE2/Ang-(1-7)/Mas axis. Results DIZE remarkably increased the expression of ACE2 and inhibited the expression of IL-6, IL-8, and MCP-1 at both mRNA and protein levels in both RPE cell lines stimulated with LPS. Inhibitors of p38, ERK1/2, JNK, and NF-κB significantly decreased LPS-induced overproduction of IL-6, IL-8, and MCP-1. DIZE reduced the expression of Ang II and AT1R, whereas increased Ang-(1-7). Furthermore, DIZE downregulated the phosphorylation of p38MAPK, ERK1/2, JNK, and the activation of NF-κB upon stimulation with LPS. Downregulating ACE2 and pre-treatment with A779 abrogated the effects of DIZE on production of cytokines, the expression of Ang II, Ang-(1-7), AT1R, phosphorylation of MAPKs and activation of NF-κB. Conclusions DIZE inhibits LPS-induced inflammatory response by activating ACE2/Ang-(1-7)/Mas axis in human RPE cells. The protective effect is mediated by inhibiting the p38MAPK, ERK1/2, JNK, and NF-κB pathways.
Collapse
Affiliation(s)
- Lifei Tao
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| | - Yiguo Qiu
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| | - Xinyu Fu
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| | - Ru Lin
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| | - Chunyan Lei
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| | - Jiaming Wang
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| | - Bo Lei
- Department of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, 1 You Yi Road, Yu Zhong District, Chongqing, 400016, China.
| |
Collapse
|
35
|
Diminazene aceturate—An antiparasitic drug of antiquity: Advances in pharmacology & therapeutics. Pharmacol Res 2015; 102:138-57. [DOI: 10.1016/j.phrs.2015.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/28/2015] [Accepted: 10/09/2015] [Indexed: 12/31/2022]
|
36
|
Diminazene aceturate (Berenil), a new use for an old compound? Int Immunopharmacol 2014; 21:342-5. [DOI: 10.1016/j.intimp.2014.05.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/21/2014] [Accepted: 05/22/2014] [Indexed: 11/20/2022]
|
37
|
Qiu Y, Shil PK, Zhu P, Yang H, Verma A, Lei B, Li Q. Angiotensin-converting enzyme 2 (ACE2) activator diminazene aceturate ameliorates endotoxin-induced uveitis in mice. Invest Ophthalmol Vis Sci 2014; 55:3809-18. [PMID: 24854854 DOI: 10.1167/iovs.14-13883] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PURPOSE Uveitis is a common cause of vision loss. The renin angiotensin system (RAS), which plays a vital role in cardiovascular system, is a potent mediator of inflammation and has been implicated in the pathogenesis of uveitis. A newly identified axis of RAS, ACE2/Ang-(1-7)/Mas, has emerged as a novel target because it counteracts the deleterious effect of angiotensin II. The purpose of this study was to investigate the effect of endogenous ACE2 activation in preventing endotoxin-induced uveitis (EIU) in mice. METHODS ACE2 activator diminazene aceturate (DIZE) was administered both systemically and locally. For systemic administration, female BALB/c mice received intraperitoneal injection of DIZE (60 mg/kg body weight [BW]) for 2 days prior to lipopolysaccharide (LPS) intravitreal injection (125 ng) to induce uveitis. For local study, DIZE was given at 0.5, 0.1, and 0 mg/mL as eyedrops six times per day for 2 days before LPS injection. The anterior segment of the mice was examined at 12, 24, 48, and 72 hours after LPS injection, and clinical scores were determined at the same time. Morphology and infiltrating inflammatory cells were evaluated after 24 hours. The mRNA levels of inflammatory cytokines were analyzed by real-time RT-PCR. ACE2 activity was determined using a self-quenching fluorescent substrate. RESULTS At 24 hours, the clinical score of mice treated with DIZE systemically was significantly lower (mean, ∼1.75) than the saline vehicle group (mean, ∼4) (P < 0.001). Histological examination showed 63.4% reduction of infiltrating inflammatory cells in the anterior segment and 57.4% reduction in the posterior segment of DIZE-treated eyes. The number of CD45(+) inflammatory cells in the vitreous of the DIZE-treated group was decreased (43.3%) compared to the vehicle group (P < 0.01). The mRNA levels of inflammatory cytokines were significantly reduced in the DIZE-treated group (P < 0.01, P < 0.001). The number of infiltrating inflammatory cells was also significantly reduced in eyes that received topical administration of DIZE: 73.8% reduction in the 0.5 mg/mL group and 51.7% reduction in the 0.1mg/mL group compared to the control group. DIZE treatment resulted in significantly increased ACE2 activity in the retina (P < 0.001). CONCLUSIONS Endogenous ACE2 activation by DIZE has a preventive effect on LPS-induced ocular inflammation in the EIU mouse model. These results support the notions that RAS plays a role in modulating ocular immune response and that enhancing ACE2 provides a novel therapeutic strategy for uveitis.
Collapse
Affiliation(s)
- Yiguo Qiu
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Pollob Kumar Shil
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States
| | - Ping Zhu
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States
| | - Hongxia Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Amrisha Verma
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States
| | - Bo Lei
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Qiuhong Li
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States
| |
Collapse
|